US10875860B2 - Azaindenoisoquinoline compounds and uses thereof - Google Patents

Azaindenoisoquinoline compounds and uses thereof Download PDF

Info

Publication number
US10875860B2
US10875860B2 US16/471,945 US201716471945A US10875860B2 US 10875860 B2 US10875860 B2 US 10875860B2 US 201716471945 A US201716471945 A US 201716471945A US 10875860 B2 US10875860 B2 US 10875860B2
Authority
US
United States
Prior art keywords
cancer
compound
mmol
compounds
optionally substituted
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
US16/471,945
Other versions
US20200095243A1 (en
Inventor
Mark S. Cushman
Ping Wang
Yves George Pommier
Mohamed S. A. Elsayed
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
US Department of Health and Human Services
Purdue Research Foundation
Original Assignee
Purdue Research Foundation
US Department of Health and Human Services
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Purdue Research Foundation, US Department of Health and Human Services filed Critical Purdue Research Foundation
Priority to US16/471,945 priority Critical patent/US10875860B2/en
Assigned to PURDUE RESEARCH FOUNDATION reassignment PURDUE RESEARCH FOUNDATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: WANG, PING, CUSHMAN, MARK S, ELSAYED, MOHAMED S.A.
Assigned to PURDUE RESEARCH FOUNDATION reassignment PURDUE RESEARCH FOUNDATION ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: WANG, PING, CUSHMAN, MARK S., ELSAYED, MOHAMED S.A.
Publication of US20200095243A1 publication Critical patent/US20200095243A1/en
Assigned to THE UNITED STATES OF AMERICA, AS REPRESENTED BY THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES reassignment THE UNITED STATES OF AMERICA, AS REPRESENTED BY THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: POMMIER, YVES G.
Application granted granted Critical
Publication of US10875860B2 publication Critical patent/US10875860B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca

Definitions

  • the present disclosure generally relates to novel compounds for a variety of therapeutic uses.
  • this disclosure relates to azaindenoisoquinoline compounds as triple inhibitors of Topoisomerase 1 (Top1), and Tyrosyl-DNA Phosphodiesterases 1 and 2 (Tdp1 and Tdp2) that are useful for treatment of a cancer.
  • Top1 Topoisomerase 1
  • Tdp1 and Tdp2 Tyrosyl-DNA Phosphodiesterases 1 and 2
  • Cancer is a group of diseases involving abnormal cell growth with the potential to invade or spread to other parts of the body. There are more than 100 types of cancer that affect human beings. In 2016, there were an estimated 1,685,210 new cancer cases diagnosed and 595,690 cancer deaths in the U.S. alone ( Cancer Statistics 2016—American Cancer Society, Inc.). There are unmet and increasing needs for new and novel therapies for fighting cancers.
  • Top1 DNA topoisomerase I
  • Camptothecins (CPTs) and indenoisoquinolines are two established classes of Top1 inhibitors (Staker, et al., J. Med. Chem. 2005, 48, 2336-2345; Pommier, et al, Mol. Cancer Ther. 2009, 8, 1008-1014)).
  • the CPTs and indenoisoquinolines act through stabilization of the DNA-Top1 covalent cleavage complex (Top1cc) by a dual DNA intercalation and protein binding mechanism that leads to inhibition of the DNA re-ligation process (Pmmier, et al., Nat. Rev.
  • Tdp1 tyrosyl-DNA phosphodiesterase 1
  • Tdp1 is a member of the phospholipase D superfamily of enzymes that catalyze the hydrolysis of the 3′-phosphotyrosyl linker found in the Top1cc and other 3′-end DNA blocking lesions (Interthal, et al., Proc. Natl. Acad. Sci. U.S.A. 2001, 98, 12009-12014; Murai, et al., J. Biol. Chem. 2012, 287, 12848-12857).
  • Tdp2 tyrosyl-DNA phosphodiesterase II
  • Top2 topoisomerase II
  • Inhibition of Tdp2 may therefore be a useful approach to overcome intrinsic or acquired resistance to Top2-targeted drug therapy (Zeng, et al., J. Biol. Chem.
  • Tdp2 also promotes repair of Top1-mediated DNA damage in the absence of Tdp1 and that cells lacking both Tdp1 and Tdp2 are more sensitive to Top1 inhibitors than Tdp1-deficient cells (Maede, et al., Mol. Cancer Ther. 2014, 13, 214-220).
  • azaindenoisoquinoline compounds Described herein are azaindenoisoquinoline compounds.
  • the compounds described herein may be useful for treating a cancer.
  • those azaindenoisoquinoline compounds may be useful as triple inhibitors of Topoisomerase 1 (Top1), and Tyrosyl-DNA Phosphodiesterases 1 and 2 (Tdp1 and Tdp2) that are useful for treatment of a cancer.
  • Top1 Topoisomerase 1
  • Tdp1 and Tdp2 Tyrosyl-DNA Phosphodiesterases 1 and 2
  • compositions of such compounds are also described herein are pharmaceutical compositions of such compounds and methods for treating cancer by administering therapeutically effective amounts of such compound alone or in pharmaceutical compositions.
  • azaindenoisoquinoline compounds having the formula (I)
  • R 1 or R 3 is not hydrogen
  • azaindenoisoquinoline compounds having the formula (I), wherein R 2 is a substituent selected from the group consisting of:
  • a pharmaceutical composition comprising a compound having the formula (I), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, and a pharmaceutically acceptable carrier, diluent, and excipient.
  • described herein is a method for treating a patient with a cancer, the method comprising the step of administering a therapeutically effective amount of a compound having the formula (I), or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, diluent, and excipient to the patient in need of relief from said cancer.
  • described herein are substances for treating a patient with a cancer, the substance having the formula (I), or a pharmaceutically acceptable salt thereof, together with a pharmaceutically acceptable carrier, diluent, and excipient to the patient in need of relief from said cancer.
  • compositions containing one or more of the compounds are also described herein.
  • the compositions include a therapeutically effective amount of the one or more compounds for treating a cancer patient.
  • the compositions may include other component and/or ingredients, including, but not limited to, other therapeutically active compounds, and/or one or more pharmaceutically acceptable carriers, diluents, excipients, and the like.
  • the compounds described herein may be used alone or in combination with other compounds useful for treating cancer, including those compounds that may be therapeutically effective by the same or different modes of action.
  • the compounds described herein may be used in combination with other compounds that are administered to treat other symptoms of cancer, such as compounds administered to relieve nausea, vomiting, pain, osteoporosis, and the like.
  • a “halogen” designates F, Cl, Br or I.
  • a “halogen-substitution” or “halo” substitution designates replacement of one or more hydrogen atoms with F, Cl, Br or I.
  • alkyl refers to a saturated monovalent chain of carbon atoms, which may be optionally branched. It is understood that in embodiments that include alkyl, illustrative variations of those embodiments include lower alkyl, such as C 1 -C 6 alkyl, methyl, ethyl, propyl, 3-methylpentyl, and the like.
  • alkenyl refers to an unsaturated monovalent chain of carbon atoms including at least one double bond, which may be optionally branched. It is understood that in embodiments that include alkenyl, illustrative variations of those embodiments include lower alkenyl, such as C 2 -C 6 , C 2 -C 4 alkenyl, and the like.
  • alkynyl refers to an unsaturated monovalent chain of carbon atoms including at least one triple bond, which may be optionally branched. It is understood that in embodiments that include alkynyl, illustrative variations of those embodiments include lower alkynyl, such as C 2 -C 6 , C 2 -C 4 alkynyl, and the like.
  • cycloalkyl refers to a monovalent chain of carbon atoms, a portion of which forms a ring. It is understood that in embodiments that include cycloalkyl, illustrative variations of those embodiments include lower cycloalkyl, such as C 3 -C 8 cycloalkyl, cyclopropyl, cyclohexyl, 3-ethylcyclopentyl, and the like.
  • cycloalkenyl refers to an unsaturated monovalent chain of carbon atoms, a portion of which forms a ring. It is understood that in embodiments that include cycloalkenyl, illustrative variations of those embodiments include lower cycloalkenyl, such as C 3 -C 8 , C 3 -C 6 cycloalkenyl.
  • alkylene refers to a saturated bivalent chain of carbon atoms, which may be optionally branched. It is understood that in embodiments that include alkylene, illustrative variations of those embodiments include lower alkylene, such as C 2 -C 4 , alkylene, methylene, ethylene, propylene, 3-methylpentylene, and the like.
  • heterocyclic or “heterocycle” refers to a monovalent chain of carbon and heteroatoms, wherein the heteroatoms are selected from nitrogen, oxygen, and sulfur, and a portion of which, at least one heteroatom, forms a ring.
  • heterocycle may include both “aromatic heterocycles” and “non-aromatic heterocycles.”
  • Heterocycles include 4-7 membered monocyclic and 8-12 membered bicyclic rings, such as imidazolyl, thiazolyl, oxazolyl, oxazinyl, thiazinyl, dithianyl, dioxanyl, isoxazolyl, isothiazolyl, triazolyl, furanyl, tetrahydrofuranyl, dihydrofuranyl, pyranyl, tetrazolyl, pyrazolyl, pyrazinyl, pyridazinyl, imidazolyl, pyridinyl, pyrrolyl, dihydropyrrolyl, pyrrolidinyl, piperidinyl, piperazinyl, pyrimidinyl, morpholinyl, tetrahydrothiophenyl, thiopheny
  • aryl includes monocyclic and polycyclic aromatic carbocyclic groups, each of which may be optionally substituted.
  • optionally substituted aryl refers to an aromatic mono or polycyclic ring of carbon atoms, such as phenyl, naphthyl, and the like, which may be optionally substituted with one or more independently selected substituents, such as halo, hydroxyl, amino, alkyl, or alkoxy, alkylsulfony, cyano, nitro, and the like.
  • heteroaryl or “aromatic heterocycle” includes substituted or unsubstituted aromatic single ring structures, preferably 5- to 7-membered rings, more preferably 5- to 6-membered rings, whose ring structures include at least one heteroatom, preferably one to four heteroatoms, more preferably one or two heteroatoms.
  • heteroaryl may also include ring systems having one or two rings wherein at least one of the rings is heteroaromatic, e.g., the other cyclic rings can be cycloalkyl, cycloalkenyl, cycloalkynyl, aromatic carbocycle, heteroaryl, and/or heterocycle.
  • Heteroaryl groups include, for example, pyrrole, furan, thiophene, imidazole, oxazole, thiazole, pyrazole, pyridine, pyrazine, pyridazine, and pyrimidine.
  • each of alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkylene, and heterocycle may be optionally substituted with independently selected groups such as alkyl, haloalkyl, hydroxyalkyl, aminoalkyl, carboxylic acid and derivatives thereof, including esters, amides, and nitrites, hydroxy, alkoxy, acyloxy, amino, alky and dialkylamino, acylamino, thio, and the like, and combinations thereof.
  • substituents means that the groups in question are either unsubstituted or substituted with one or more of the substituents specified. When the groups in question are substituted with more than one substituent, the substituents may be the same or different. Furthermore, when using the terms “independently,” “independently are,” and “independently selected from” mean that the groups in question may be the same or different. Certain of the herein defined terms may occur more than once in the structure, and upon such occurrence each term shall be defined independently of the other.
  • the term “patient” includes human and non-human animals such as companion animals (dogs and cats and the like) and livestock animals. Livestock animals are animals raised for food production.
  • the patient to be treated is preferably a mammal, in particular a human being.
  • pharmaceutically acceptable carrier refers to a pharmaceutically-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting any subject composition or component thereof.
  • a pharmaceutically-acceptable material such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting any subject composition or component thereof.
  • Each carrier must be “acceptable” in the sense of being compatible with the subject composition and its components and not injurious to the patient.
  • materials which may serve as pharmaceutically acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide;
  • administering includes all means of introducing the compounds and compositions described herein to the patient, including, but are not limited to, oral (po), intravenous (iv), intramuscular (im), subcutaneous (sc), transdermal, inhalation, buccal, ocular, sublingual, vaginal, rectal, and the like.
  • the compounds and compositions described herein may be administered in unit dosage forms and/or formulations containing conventional nontoxic pharmaceutically acceptable carriers, adjuvants, and vehicles.
  • the total daily usage of the compounds and compositions described herein may be decided by the attending physician within the scope of sound medical judgment.
  • the specific therapeutically effective dose level for any particular patient will depend upon a variety of factors, including the disorder being treated and the severity of the disorder; activity of the specific compound employed; the specific composition employed; the age, body weight, general health, gender, and diet of the patient: the time of administration, and rate of excretion of the specific compound employed, the duration of the treatment, the drugs used in combination or coincidentally with the specific compound employed; and like factors well known to the researcher, veterinarian, medical doctor or other clinician of ordinary skill.
  • a wide range of permissible dosages are contemplated herein, including doses falling in the range from about 1 ⁇ g/kg to about 1 g/kg.
  • the dosage may be single or divided, and may be administered according to a wide variety of dosing protocols, including q.d., b.i.d., t.i.d., or even every other day, once a week, once a month, and the like.
  • the therapeutically effective amount described herein corresponds to the instance of administration, or alternatively to the total daily, weekly, or monthly dose.
  • the term “therapeutically effective amount” refers to that amount of active compound or pharmaceutical agent that elicits the biological or medicinal response in a tissue system, animal or human that is being sought by a researcher, veterinarian, medical doctor or other clinicians, which includes alleviation of the symptoms of the disease or disorder being treated.
  • the therapeutically effective amount is that which may treat or alleviate the disease or symptoms of the disease at a reasonable benefit/risk ratio applicable to any medical treatment.
  • the term “therapeutically effective amount” refers to the amount to be administered to a patient, and may be based on body surface area, patient weight, and/or patient condition.
  • body surface area may be approximately determined from patient height and weight (see, e.g., Scientific Tables 1970, Geigy Pharmaceuticals, Ardley, N.Y., pages 537-538).
  • a therapeutically effective amount of the compounds described herein may be defined as any amount useful for inhibiting the growth of (or killing) a population of malignant cells or cancer cells, such as may be found in a patient in need of relief from such cancer or malignancy.
  • effective amounts range from about 5 mg/kg to about 500 mg/kg, from about 5 mg/kg to about 250 mg/kg, and/or from about 5 mg/kg to about 150 mg/kg of compound per patient body weight. It is appreciated that effective doses may also vary depending on the route of administration, optional excipient usage, and the possibility of co-usage of the compound with other conventional and non-conventional therapeutic treatments, including other anti-tumor agents, radiation therapy, and the like.
  • the term “about” can allow for a degree of variability in a value or range, for example, within 10%, within 5%, or within 1% of a stated value or of a stated limit of a range.
  • the term “substantially” can allow for a degree of variability in a value or range, for example, within 90%, within 95%, or within 99% of a stated value or of a stated limit of a range.
  • the present invention relates to azaindenoisoquinoline compounds having the formula (I)
  • R 1 or R 3 is not hydrogen
  • the present invention relates to azaindenoisoquinoline compounds having the formula (I), wherein one of R 1 is chloro or fluoro and the remaining substituents are hydrogen.
  • the present invention relates to azaindenoisoquinoline compounds having the formula (I), wherein one of R 1 is nitro and the remaining substituents are hydrogen.
  • the present invention relates to azaindenoisoquinoline compounds having the formula (I), wherein R 2 is a halo or cyano.
  • the present invention related to azaindenoisoquinoline compounds having the formula (I), wherein R 2 is a substituent selected from the group consisting of:
  • azaindenoisoquinoline compounds having the formula (II)
  • the present invention relates to azaindenoisoquinoline compounds having the formula (II), wherein one of R 1 is chloro or fluoro and the remaining substituents are hydrogen.
  • the present invention relates to azaindenoisoquinoline compounds having the formula (II), wherein one of R 1 is nitro and the remaining substituents are hydrogen.
  • the present invention relates to azaindenoisoquinoline compounds having the formula (II), wherein R 2 is a halo or cyano.
  • the present invention related to azaindenoisoquinoline compounds having the formula (II), wherein R 2 is a substituent selected from the group consisting of:
  • the present invention relates to azaindenoisoquinoline compounds having the formula (I), wherein one of R 3 is an alkoxy and the remaining R 3 substituents are hydrogen.
  • the present invention relates to azaindenoisoquinoline compounds having the formula (II), wherein one of R 3 is alkoxy and the remaining R 3 substituents are hydrogen.
  • the present invention relates a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers, diluents, and excipients.
  • the present invention relates a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula (II), or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers, diluents, and excipients.
  • the present invention relates a method for treating a patient with a cancer, comprising the step of administering a therapeutically effective amount of the pharmaceutical composition disclosed herein to the patient in need of relief from said cancer.
  • the present invention relates a method for treating a patient with a cancer, the method comprising the step of administering a therapeutically effective amount of a compound disclosed herein, together with one or more pharmaceutically acceptable carriers, diluents, and excipients, to the patient in need of relief from said cancer.
  • the present invention relates a method for treating a patient with a cancer, the method comprising the step of administering a therapeutically effective amount of a compound disclosed herein, and a therapeutically effective amount of one or more other compounds of the same or different mode of action, together one or more pharmaceutically acceptable carriers, diluents, and excipients, to the patient in need of relief from said cancer.
  • the present invention relates a method for treating a patient with a cancer, the method comprising the step of administering a therapeutically effective amount of a compound functioning as a triple inhibitor toward human topoisomerase 1, tyrosyl-DNA phosphodiesterase 1, and tyrosyl-DNA phosphodiesterase 2.
  • the present invention relates a substance for use in the treatment of a cancer, the substance having formula (I)
  • R 1 or R 3 is not hydrogen
  • the anhydrides 11a and 11b were prepared by published literature procedures (Kang, B.-R, et al., Med. Chem. Res. 2014, 23, 1340-1349). Bromination of 5-methoxy-3-methylpicolinonitrile in the presence of the radical initiator AIBN produced intermediate bromide 12a (Kiselev, et al, J. Med. Chem. 2011, 54, 6106-6116), which was used directly in the next step without additional purification. The condensation of 12a and 11a-b in acetonitrile promoted by Et 3 N afforded compounds 13a-b in ⁇ 25% yield for two steps.
  • Oxidation of 13a-b with selenium dioxide provided azaindenoisoquinoline intermediates 14a-b.
  • Treating compounds 14a-b with NaH in DMF at 0° C. followed by reaction with 1-chloro-3-bromopropane yielded the common intermediates 15a-b.
  • the common intermediates 15a-b were used for the synthesis of the final compounds 16a-j and 17a-i by alkylation of the corresponding amines in DMF as shown in Scheme 4.
  • Top1 inhibition was recorded as the ability of the drug to induce enzyme-linked DNA breaks in the Top1-mediated DNA cleavage assay (Dexheimer, et al., Nat. Protoc. 2008, 3, 1736-1750).
  • compound 11 in which a methylamine was introduced at the end of propyl chain, displayed improved Top inhibitory activity relative to 7 at the +++ level.
  • the isopropylamine 14 and ethylamine 15 compounds demonstrated excellent Top1 inhibitory activity at the ++++ level.
  • the 4-2(aminoethyl)morpholine compound 13 and 2-amino-2-thiazoline compound 12 displayed improved Top1 inhibitory activity relative to 7 at the +++ and ++++ levels, respectively.
  • the improved potencies of compounds 11-15 compared to those of 8-10 indicate that a secondary amine in side chain of 7-azaindenoisoquinolines is better than a tertiary amine for the Top1 inhibitory activity.
  • the Tdp1 inhibitory activities of the 7-azaindenoisoquinolines were determining by measuring their abilities to inhibit the hydrolysis of the phosphodiester linkage between the tyrosine residue and the 3′-end of a DNA oligonucleotide substrate (N14Y), thus preventing the generation of an oligonucleotide with a free 3′-phosphate (N14P) (Dexheimer, et al., J. Med. Chem. 2009, 52, 7122-7131).
  • the Tdp1 inhibitory activities of the 7-azaindenoisoquinolines are summarized in Table 1.
  • Tdp1 inhibitory potencies were determined in duplicate using a semiquantitative scale: 0, IC 50 >111 ⁇ M; +, IC 50 between 37 and 111 ⁇ M; ++, IC 50 between 12 and 37 ⁇ M; +++, IC 50 between 1 and 12 ⁇ M; and ++++, IC 50 ⁇ 1 ⁇ M.
  • IC 50 values were determined in duplicate using a semiquantitative scale: 0, IC 50 > 111 ⁇ M; +, IC 50 between 37 and 111 ⁇ M; ++, IC 50 between 12 and 37 ⁇ M; +++, IC 50 between 1 and 12 ⁇ M; ++++, IC 50 ⁇ 1 ⁇ M.
  • the 7-azaindenoisoquinolines 7-17 were evaluated in the National Cancer Institute's Developmental Therapeutics Program screen against 60 human cancer cell lines (the “NCI-60,” Shoemaker, R. H. Nat. Rev. Cancer 2006, 6, 813-823).
  • the GI 50 values obtained with selected cell lines, along with the mean graph midpoint (MGM) values, are summarized in Table 1.
  • the MGM was based on a calculation of the average GI 50 for all of the cell lines tested in which GI 50 values below and above the test range (0.01 ⁇ M to 100 ⁇ M) were taken as the minimum (0.01 ⁇ M) and maximum (100 ⁇ M) drug concentrations used in the screening test.
  • the Top1 ++++ compounds 12, 14, and 15 were among the most cytotoxic, with MGM values of 0.058, 0.058, and 0.079 ⁇ M, respectively. The differences in cytotoxicity among these three compounds are low despite clear differences in Tdp1 and Tdp2 inhibition, suggesting that Tdp1 and Tdp2 inhibition do not contribute significantly to the cytotoxicities of these compounds.
  • the Top1 +++ compounds are 11 (MGM 0.714 ⁇ M), 13 (MGM 0.177 ⁇ M), and 16 (MGM 1.650 ⁇ M).
  • Tdp1 and Tdp2 inhibitory potencies 16 is expected to be the least potent, which it clearly is. However, one would expect 11 to be the most cytotoxic, and in fact 13 is the most cytotoxic of the three compounds.
  • Top1++ compounds 7 (MGM 0.112 M), 8 (MGM 0.045 ⁇ M), 9 (MGM 0.562 ⁇ M), 10 (MGM 0.316 ⁇ M), and 17 (not tested due to low cytotoxicity in the one-concentration preliminary testing), 17 is expected to be the most cytotoxic on the basis of Tdp1 and Tdp2 inhibition, but it is actually the least cytotoxic.
  • compounds 7 or 10 should be the least cytotoxic, but actually 17 is.
  • compound 8 is surprisingly the most cytotoxic compound in the whole series despite having relatively moderate Top1 inhibitory activity.
  • these triple inhibitors are very cytotoxic anticancer agents, but their relative cytotoxicities cannot be rationalized simply on the basis of their inhibitory activities vs. the three enzymes.
  • Other factors that could contribute to the lack of agreement between the enzyme inhibitory activities and cytotoxicities of these 7-azaindenoisoquinoline include differences in cellular uptake, distribution within the cell, metabolism, efflux from the cell, off-target effects, and lack of sufficient potency vs. Tdp1 and Tdp2 to exert a significant synergistic effect.
  • Top1-mediated DNA cleavage assay scores the activity of Top poisons with a rubric based on the activity of 1 ⁇ M camptothecin. Test agents are incubated at 0.1, 1, 10, and 100 ⁇ M concentrations with a 3′-[ 32 P]-labeled double-stranded DNA fragment and Top1 enzyme. Top1 poisons bind to and trap Top1-DNA cleavage complexes.
  • the DNA cleavage pattern is then documented by gel electrophoresis. Visual comparison of the lanes produced with 1 ⁇ M CPT indicates the activity of the new compounds. Finally, a semiquantitative score which ranges from 0 (no activity) to ++++(activity equal to that of 1 ⁇ M CPT) is used to describe the activity of the new compounds (see Table 2 caption for a complete description of the scoring rubric).
  • the biological activities of the compounds are summarized in Table 2. The compounds have a good to moderate Top1 inhibitory activity. Of the nineteen compounds synthesized, compound 16d displayed the best activity with a score of ++++, while five other compounds exhibited good activity with a score of +++. On the other hand, five compounds showed weak activity with only a “+” score. Some of the compounds act as Top1 suppressors at high concentration, which likely results from the binding of the drug to the DNA at high drug concentration, making the DNA a poorer substrate for the cleavage reaction.
  • NMR spectra were obtained at 300 or 500 ( 1 H) and 75 or 125 ( 13 C) MHz using Bruker ARX300 or Bruker DX-2 500 [QNP probe or multinuclear broadband observe (BBO) probe, respectively] spectrometers. Column chromatography was performed with 230-400 mesh silica gel. The melting points were determined using capillary tubes with a Mel-Temp apparatus and are uncorrected. IR spectra were obtained using a Perkin-Elmer 1600 series FTIR spectrometer on salt plates or as KBr pellets. ESI-MS analyses were recorded on a FinniganMAT LCQ Classic mass spectrometer. APCI-MS analyses were performed using an Agilent 6320 ion trap mass spectrometer.
  • a 3′-[ 32 P]-labeled 117-bp DNA oligonucleotide was prepared as previously described.
  • the oligonucleotide contains previously identified Top1 cleavage sites in 161-bp pBluescript SK( ⁇ ) phagemid DNA.
  • Approximately 2 nM radiolabeled DNA substrate was incubated with recombinant Top1 in 20 ⁇ L of reaction buffer [10 mM Tris-HCl (pH 7.5), 50 mM KCl, 5 mM MgCl 2 , 0.1 mM EDTA, and 15 ⁇ g/mL BSA] at 25° C.
  • TDP2 reactions were carried out as described previously with the following modifications.
  • the 18-mer single-stranded oligonucleotide DNA substrate (TY18, ⁇ 32 P-cordycepin-3′-labeled) was incubated at 1 nM with 25 pM recombinant human TDP2 in the absence or presence of inhibitor for 15 min at room temperature in the LMP2 assay buffer containing 50 mM Tris-HCl, pH 7.5, 80 mM KCl, 5 mM MgCl 2 , 0.1 mM EDTA, 1 mM DTT, 40 ⁇ g/mL BSA, and 0.01% Tween 20. Reactions were terminated and treated similarly to whole-cell extract and recombinant Tdp1 reactions (see previous paragraph).

Abstract

Tyrosyl-DNA Phosphodiesterases 1 and 2 (Tdp1 and Tdp2) can repair damaged DNA resulting from topoisomerase inhibitors (e.g. Top1) and a variety of other DNA-damaging agents. 7-Azaindenoisoquinolines that are inhibitors of each of Top1, Tdp1 and Tdp2 are disclosed. Also described are methods for preparing azaindenoisoquinoline and methods for treating patients of a cancer using the disclosed azaindenoisoquinoline compounds or a pharmaceutical formulation thereof.

Description

CROSS-REFERENCE TO RELATED APPLICATIONS
This application is a national application under 37 C.F.R § 371(b) of International Application No. PCT/US2017/067206, filed Dec. 19, 2017, which claims the benefit of U.S. Provisional Patent Application No. 62/437,777, filed Dec. 22, 2016, the contents of each of which are hereby incorporated by reference in their entirety into this disclosure.
GOVERNMENT RIGHTS
This invention was made with government support under CA023168, awarded by the National Institutes of Health. The United States government has certain rights in the invention.
TECHNICAL FIELD
The present disclosure generally relates to novel compounds for a variety of therapeutic uses. In particular, this disclosure relates to azaindenoisoquinoline compounds as triple inhibitors of Topoisomerase 1 (Top1), and Tyrosyl-DNA Phosphodiesterases 1 and 2 (Tdp1 and Tdp2) that are useful for treatment of a cancer.
BACKGROUNDS
This section introduces aspects that may help facilitate a better understanding of the disclosure. Accordingly, these statements are to be read in this light and are not to be understood as admissions about what is or is not prior art.
Cancer is a group of diseases involving abnormal cell growth with the potential to invade or spread to other parts of the body. There are more than 100 types of cancer that affect human beings. In 2016, there were an estimated 1,685,210 new cancer cases diagnosed and 595,690 cancer deaths in the U.S. alone (Cancer Statistics 2016—American Cancer Society, Inc.). There are unmet and increasing needs for new and novel therapies for fighting cancers.
The inhibition of DNA topoisomerase I (Top1) has proven to be a successful approach to the design of anticancer agents. Camptothecins (CPTs) and indenoisoquinolines are two established classes of Top1 inhibitors (Staker, et al., J. Med. Chem. 2005, 48, 2336-2345; Pommier, et al, Mol. Cancer Ther. 2009, 8, 1008-1014)). The CPTs and indenoisoquinolines act through stabilization of the DNA-Top1 covalent cleavage complex (Top1cc) by a dual DNA intercalation and protein binding mechanism that leads to inhibition of the DNA re-ligation process (Pmmier, et al., Nat. Rev. Drug Discovery 2012, 11, 25-36). Subsequent collision of the DNA replication fork with drug-stabilized complexes causes DNA double-stranded breaks. Ultimately, this leads to tumor cell death. DNA repair after Top1-mediated DNA cleavage is a complex process that can be initiated by tyrosyl-DNA phosphodiesterase 1 (Tdp1) (Plo, et al., DNA Repair 2003, 2, 1087-1100; Davies, et al., J. Mol. Biol. 2002, 324, 917-932), which plays a critical role in development of drug resistance (Perego, et al., Biochem. Pharmacol. 2012, 83, 27-36). Tdp1 is a member of the phospholipase D superfamily of enzymes that catalyze the hydrolysis of the 3′-phosphotyrosyl linker found in the Top1cc and other 3′-end DNA blocking lesions (Interthal, et al., Proc. Natl. Acad. Sci. U.S.A. 2001, 98, 12009-12014; Murai, et al., J. Biol. Chem. 2012, 287, 12848-12857).
On the other hand, tyrosyl-DNA phosphodiesterase II (Tdp2) is a member of the metal-dependent phosphodiesterases that was recently discovered with repair function linked to topoisomerase II (Top2)-mediated DNA damage (Pommier, et al., DNA Repair 2014, 19, 114-129). Tdp2 cleaves Top2-DNA adducts by catalyzing the hydrolysis of 5′-phosphotyrosyl bonds and thereby releasing trapped Top2 from 5′-termini, thus playing a key role in maintaining normal DNA topology. Inhibition of Tdp2 may therefore be a useful approach to overcome intrinsic or acquired resistance to Top2-targeted drug therapy (Zeng, et al., J. Biol. Chem. 2011, 286, 403-409; Ledesma, et al., Nature, 2009, 461, 674). More recently, it was revealed that Tdp2 also promotes repair of Top1-mediated DNA damage in the absence of Tdp1 and that cells lacking both Tdp1 and Tdp2 are more sensitive to Top1 inhibitors than Tdp1-deficient cells (Maede, et al., Mol. Cancer Ther. 2014, 13, 214-220). The hypothesis that Tdp2 may serve as a potential therapeutic co-target of Top1 and Tdp1 needs to be investigated (Pommier, et al., 2014; Zeng, et al., 2011).
BRIEF SUMMARY OF INVENTION
Described herein are azaindenoisoquinoline compounds. The compounds described herein may be useful for treating a cancer. In particular those azaindenoisoquinoline compounds may be useful as triple inhibitors of Topoisomerase 1 (Top1), and Tyrosyl-DNA Phosphodiesterases 1 and 2 (Tdp1 and Tdp2) that are useful for treatment of a cancer.
Also described herein are pharmaceutical compositions of such compounds and methods for treating cancer by administering therapeutically effective amounts of such compound alone or in pharmaceutical compositions.
In some illustrative embodiments, described herein are azaindenoisoquinoline compounds having the formula (I)
Figure US10875860-20201229-C00001

or a pharmaceutically acceptable salt thereof, wherein:
    • m is an integer from 1 to about 6;
  • R1 represents 1-4 substituents each of which is independently selected from the group consisting of hydrogen, halo, hydroxy, nitro, cyano, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkylthio, optionally substituted alkylsulfonyl, carboxylic acid, and sulfonic acid; or R1 represents 2-4 substituents where 2 of said substituents are adjacent substituents and are taken together with the attached carbons to form an optionally substituted heterocycle, and where any remaining substituents are each independently selected from the group consisting of hydrogen, halo, hydroxy, optionally substituted alkyl, optionally substituted alkoxy, cyano, nitro, optionally substituted alkylthio, optionally substituted alkylsulfonyl, carboxylic acid, and sulfonic acid;
  • R2 is selected from the group consisting of heteroaryl, heteroaryloxy, heteroarylamino, heteroarylalkylaminoalkylamino, heterocyclyl, heterocyclylamino, amino, hydroxyl, halo, cyano, alkylamino, dialkylamino, trialkylammonium, bis(hydroxyalkyl)amino, and hydroxyalkylaminoalkylamino, wherein each of heteroaryl, heteroaryloxy, and heteroarylamino, heteroarylalkylaminoalkylamino, heterocyclyl, and heterocyclylamino is optionally substituted; and
  • R3 represents 1-3 substituents each of which is independently selected from the group consisting of hydrogen, halo, hydroxy, cyano, nitro, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkylthio, optionally substituted alkylsulfonyl, carboxylic acid, and sulfonic acid; or R3 represents 2-3 substituents where 2 of said substituents are adjacent substituents and are taken together with the attached carbons to form an optionally substituted heterocycle, and where the remaining substituent is independently selected from the group consisting of hydrogen, halo, hydroxy, optionally substituted alkyl, optionally substituted alkoxy, cyano, nitro, optionally substituted alkylthio, optionally substituted alkylsulfonyl, carboxylic acid, and sulfonic acid, and
wherein at least one of R1 or R3 is not hydrogen.
In some other embodiments, described herein are azaindenoisoquinoline compounds having the formula (I), wherein R2 is a substituent selected from the group consisting of:
Figure US10875860-20201229-C00002
In some illustrative embodiments, described herein is a pharmaceutical composition comprising a compound having the formula (I), or a pharmaceutically acceptable salt, hydrate, or solvate thereof, and a pharmaceutically acceptable carrier, diluent, and excipient.
In some other illustrative embodiments, described herein is a method for treating a patient with a cancer, the method comprising the step of administering a therapeutically effective amount of a compound having the formula (I), or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable carrier, diluent, and excipient to the patient in need of relief from said cancer.
In some other illustrative embodiments, described herein are substances for treating a patient with a cancer, the substance having the formula (I), or a pharmaceutically acceptable salt thereof, together with a pharmaceutically acceptable carrier, diluent, and excipient to the patient in need of relief from said cancer.
In some embodiments, pharmaceutical compositions containing one or more of the compounds are also described herein. In one aspect, the compositions include a therapeutically effective amount of the one or more compounds for treating a cancer patient. It is to be understood that the compositions may include other component and/or ingredients, including, but not limited to, other therapeutically active compounds, and/or one or more pharmaceutically acceptable carriers, diluents, excipients, and the like.
It is appreciated herein that the compounds described herein may be used alone or in combination with other compounds useful for treating cancer, including those compounds that may be therapeutically effective by the same or different modes of action. In addition, it is appreciated herein that the compounds described herein may be used in combination with other compounds that are administered to treat other symptoms of cancer, such as compounds administered to relieve nausea, vomiting, pain, osteoporosis, and the like.
DETAILED DESCRIPTION
For the purposes of promoting an understanding of the principles of the present disclosure, references will now be made to the embodiments illustrated in the drawings, and specific language will be used to describe the same. It will nevertheless be understood that no limitation of the scope of this disclosure is thereby intended.
As used herein, the following terms and phrases shall have the meanings set forth below. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood to one of ordinary skill in the art.
A “halogen” designates F, Cl, Br or I. A “halogen-substitution” or “halo” substitution designates replacement of one or more hydrogen atoms with F, Cl, Br or I.
As used herein, the term “alkyl” refers to a saturated monovalent chain of carbon atoms, which may be optionally branched. It is understood that in embodiments that include alkyl, illustrative variations of those embodiments include lower alkyl, such as C1-C6 alkyl, methyl, ethyl, propyl, 3-methylpentyl, and the like.
As used herein, the term “alkenyl” refers to an unsaturated monovalent chain of carbon atoms including at least one double bond, which may be optionally branched. It is understood that in embodiments that include alkenyl, illustrative variations of those embodiments include lower alkenyl, such as C2-C6, C2-C4 alkenyl, and the like.
As used herein, the term “alkynyl” refers to an unsaturated monovalent chain of carbon atoms including at least one triple bond, which may be optionally branched. It is understood that in embodiments that include alkynyl, illustrative variations of those embodiments include lower alkynyl, such as C2-C6, C2-C4 alkynyl, and the like.
As used herein, the term “cycloalkyl” refers to a monovalent chain of carbon atoms, a portion of which forms a ring. It is understood that in embodiments that include cycloalkyl, illustrative variations of those embodiments include lower cycloalkyl, such as C3-C8 cycloalkyl, cyclopropyl, cyclohexyl, 3-ethylcyclopentyl, and the like.
As used herein, the term “cycloalkenyl” refers to an unsaturated monovalent chain of carbon atoms, a portion of which forms a ring. It is understood that in embodiments that include cycloalkenyl, illustrative variations of those embodiments include lower cycloalkenyl, such as C3-C8, C3-C6 cycloalkenyl.
As used herein, the term “alkylene” refers to a saturated bivalent chain of carbon atoms, which may be optionally branched. It is understood that in embodiments that include alkylene, illustrative variations of those embodiments include lower alkylene, such as C2-C4, alkylene, methylene, ethylene, propylene, 3-methylpentylene, and the like.
As used herein, the term “heterocyclic” or “heterocycle” refers to a monovalent chain of carbon and heteroatoms, wherein the heteroatoms are selected from nitrogen, oxygen, and sulfur, and a portion of which, at least one heteroatom, forms a ring. The term “heterocycle” may include both “aromatic heterocycles” and “non-aromatic heterocycles.” Heterocycles include 4-7 membered monocyclic and 8-12 membered bicyclic rings, such as imidazolyl, thiazolyl, oxazolyl, oxazinyl, thiazinyl, dithianyl, dioxanyl, isoxazolyl, isothiazolyl, triazolyl, furanyl, tetrahydrofuranyl, dihydrofuranyl, pyranyl, tetrazolyl, pyrazolyl, pyrazinyl, pyridazinyl, imidazolyl, pyridinyl, pyrrolyl, dihydropyrrolyl, pyrrolidinyl, piperidinyl, piperazinyl, pyrimidinyl, morpholinyl, tetrahydrothiophenyl, thiophenyl, azetidinyl, oxetanyl, thiiranyl, oxiranyl, aziridinyl, and the like. “Heterocycles” may be optionally substituted at any one or more positions capable of bearing a hydrogen atom.
As used herein, the term “aryl” includes monocyclic and polycyclic aromatic carbocyclic groups, each of which may be optionally substituted. The term “optionally substituted aryl” refers to an aromatic mono or polycyclic ring of carbon atoms, such as phenyl, naphthyl, and the like, which may be optionally substituted with one or more independently selected substituents, such as halo, hydroxyl, amino, alkyl, or alkoxy, alkylsulfony, cyano, nitro, and the like.
The term “heteroaryl” or “aromatic heterocycle” includes substituted or unsubstituted aromatic single ring structures, preferably 5- to 7-membered rings, more preferably 5- to 6-membered rings, whose ring structures include at least one heteroatom, preferably one to four heteroatoms, more preferably one or two heteroatoms. The term “heteroaryl” may also include ring systems having one or two rings wherein at least one of the rings is heteroaromatic, e.g., the other cyclic rings can be cycloalkyl, cycloalkenyl, cycloalkynyl, aromatic carbocycle, heteroaryl, and/or heterocycle. Heteroaryl groups include, for example, pyrrole, furan, thiophene, imidazole, oxazole, thiazole, pyrazole, pyridine, pyrazine, pyridazine, and pyrimidine.
It is understood that each of alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkylene, and heterocycle may be optionally substituted with independently selected groups such as alkyl, haloalkyl, hydroxyalkyl, aminoalkyl, carboxylic acid and derivatives thereof, including esters, amides, and nitrites, hydroxy, alkoxy, acyloxy, amino, alky and dialkylamino, acylamino, thio, and the like, and combinations thereof.
The term “optionally substituted,” or “optional substituents,” as used herein, means that the groups in question are either unsubstituted or substituted with one or more of the substituents specified. When the groups in question are substituted with more than one substituent, the substituents may be the same or different. Furthermore, when using the terms “independently,” “independently are,” and “independently selected from” mean that the groups in question may be the same or different. Certain of the herein defined terms may occur more than once in the structure, and upon such occurrence each term shall be defined independently of the other.
The term “patient” includes human and non-human animals such as companion animals (dogs and cats and the like) and livestock animals. Livestock animals are animals raised for food production. The patient to be treated is preferably a mammal, in particular a human being.
The term “pharmaceutically acceptable carrier” is art-recognized and refers to a pharmaceutically-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting any subject composition or component thereof. Each carrier must be “acceptable” in the sense of being compatible with the subject composition and its components and not injurious to the patient. Some examples of materials which may serve as pharmaceutically acceptable carriers include: (1) sugars, such as lactose, glucose and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide; (15) alginic acid; (16) pyrogen-free water; (17) isotonic saline; (18) Ringer's solution; (19) ethyl alcohol; (20) phosphate buffer solutions; and (21) other non-toxic compatible substances employed in pharmaceutical formulations.
As used herein, the term “administering” includes all means of introducing the compounds and compositions described herein to the patient, including, but are not limited to, oral (po), intravenous (iv), intramuscular (im), subcutaneous (sc), transdermal, inhalation, buccal, ocular, sublingual, vaginal, rectal, and the like. The compounds and compositions described herein may be administered in unit dosage forms and/or formulations containing conventional nontoxic pharmaceutically acceptable carriers, adjuvants, and vehicles.
It is to be understood that the total daily usage of the compounds and compositions described herein may be decided by the attending physician within the scope of sound medical judgment. The specific therapeutically effective dose level for any particular patient will depend upon a variety of factors, including the disorder being treated and the severity of the disorder; activity of the specific compound employed; the specific composition employed; the age, body weight, general health, gender, and diet of the patient: the time of administration, and rate of excretion of the specific compound employed, the duration of the treatment, the drugs used in combination or coincidentally with the specific compound employed; and like factors well known to the researcher, veterinarian, medical doctor or other clinician of ordinary skill.
Depending upon the route of administration, a wide range of permissible dosages are contemplated herein, including doses falling in the range from about 1 μg/kg to about 1 g/kg. The dosage may be single or divided, and may be administered according to a wide variety of dosing protocols, including q.d., b.i.d., t.i.d., or even every other day, once a week, once a month, and the like. In each case the therapeutically effective amount described herein corresponds to the instance of administration, or alternatively to the total daily, weekly, or monthly dose.
As used herein, the term “therapeutically effective amount” refers to that amount of active compound or pharmaceutical agent that elicits the biological or medicinal response in a tissue system, animal or human that is being sought by a researcher, veterinarian, medical doctor or other clinicians, which includes alleviation of the symptoms of the disease or disorder being treated. In one aspect, the therapeutically effective amount is that which may treat or alleviate the disease or symptoms of the disease at a reasonable benefit/risk ratio applicable to any medical treatment.
As used herein, the term “therapeutically effective amount” refers to the amount to be administered to a patient, and may be based on body surface area, patient weight, and/or patient condition. In addition, it is appreciated that there is an interrelationship of dosages determined for humans and those dosages determined for animals, including test animals (illustratively based on milligrams per meter squared of body surface) as described by Freireich, E. J., et al., Cancer Chemother. Rep. 1966, 50 (4), 219, the disclosure of which is incorporated herein by reference. Body surface area may be approximately determined from patient height and weight (see, e.g., Scientific Tables 1970, Geigy Pharmaceuticals, Ardley, N.Y., pages 537-538). A therapeutically effective amount of the compounds described herein may be defined as any amount useful for inhibiting the growth of (or killing) a population of malignant cells or cancer cells, such as may be found in a patient in need of relief from such cancer or malignancy. Typically, such effective amounts range from about 5 mg/kg to about 500 mg/kg, from about 5 mg/kg to about 250 mg/kg, and/or from about 5 mg/kg to about 150 mg/kg of compound per patient body weight. It is appreciated that effective doses may also vary depending on the route of administration, optional excipient usage, and the possibility of co-usage of the compound with other conventional and non-conventional therapeutic treatments, including other anti-tumor agents, radiation therapy, and the like.
In the present disclosure the term “about” can allow for a degree of variability in a value or range, for example, within 10%, within 5%, or within 1% of a stated value or of a stated limit of a range. In the present disclosure the term “substantially” can allow for a degree of variability in a value or range, for example, within 90%, within 95%, or within 99% of a stated value or of a stated limit of a range.
In some illustrative embodiments, the present invention relates to azaindenoisoquinoline compounds having the formula (I)
Figure US10875860-20201229-C00003

or a pharmaceutically acceptable salt thereof, wherein:
    • m is an integer from 1 to about 6;
  • R1 represents 1-4 substituents each of which is independently selected from the group consisting of hydrogen, halo, hydroxy, nitro, cyano, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkylthio, optionally substituted alkylsulfonyl, carboxylic acid, and sulfonic acid; or R1 represents 2-4 substituents where 2 of said substituents are adjacent substituents and are taken together with the attached carbons to form an optionally substituted heterocycle, and where any remaining substituents are each independently selected from the group consisting of hydrogen, halo, hydroxy, optionally substituted alkyl, optionally substituted alkoxy, cyano, nitro, optionally substituted alkylthio, optionally substituted alkylsulfonyl, carboxylic acid, and sulfonic acid;
  • R2 is selected from the group consisting of heteroaryl, heteroaryloxy, heteroarylamino, heteroarylalkylaminoalkylamino, heterocyclyl, heterocyclylamino, amino, hydroxyl, halo, cyano, alkylamino, dialkylamino, trialkylammonium, bis(hydroxyalkyl)amino, and hydroxyalkylaminoalkylamino, wherein each of heteroaryl, heteroaryloxy, and heteroarylamino, heteroarylalkylaminoalkylamino, heterocyclyl, and heterocyclylamino is optionally substituted; and
  • R3 represents 1-3 substituents each of which is independently selected from the group consisting of hydrogen, halo, hydroxy, cyano, nitro, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkylthio, optionally substituted alkylsulfonyl, carboxylic acid, and sulfonic acid; or R3 represents 2-3 substituents where 2 of said substituents are adjacent substituents and are taken together with the attached carbons to form an optionally substituted heterocycle, and where the remaining substituent is independently selected from the group consisting of hydrogen, halo, hydroxy, optionally substituted alkyl, optionally substituted alkoxy, cyano, nitro, optionally substituted alkylthio, optionally substituted alkylsulfonyl, carboxylic acid, and sulfonic acid, and
wherein at least one of R1 or R3 is not hydrogen.
In some other embodiments, the present invention relates to azaindenoisoquinoline compounds having the formula (I), wherein one of R1 is chloro or fluoro and the remaining substituents are hydrogen.
In some other embodiments, the present invention relates to azaindenoisoquinoline compounds having the formula (I), wherein one of R1 is nitro and the remaining substituents are hydrogen.
In some other embodiments, the present invention relates to azaindenoisoquinoline compounds having the formula (I), wherein R2 is a halo or cyano.
In some other embodiments, the present invention related to azaindenoisoquinoline compounds having the formula (I), wherein R2 is a substituent selected from the group consisting of:
Figure US10875860-20201229-C00004
In some illustrative embodiments, described herein are azaindenoisoquinoline compounds having the formula (II)
Figure US10875860-20201229-C00005

or a pharmaceutically acceptable salt thereof, wherein:
    • m is an integer from 1 to about 6;
  • R1 represents 1-4 substituents each of which is independently selected from the group consisting of hydrogen, halo, hydroxy, nitro, cyano, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkylthio, optionally substituted alkylsulfonyl, carboxylic acid, and sulfonic acid; or R1 represents 2-4 substituents where 2 of said substituents are adjacent substituents and are taken together with the attached carbons to form an optionally substituted heterocycle, and where any remaining substituents are each independently selected from the group consisting of hydrogen, halo, hydroxy, optionally substituted alkyl, optionally substituted alkoxy, cyano, nitro, optionally substituted alkylthio, optionally substituted alkylsulfonyl, carboxylic acid, and sulfonic acid; and
  • R2 is selected from the group consisting of heteroaryl, heteroaryloxy, heteroarylamino, heteroarylalkylaminoalkylamino, heterocyclyl, heterocyclylamino, amino, hydroxyl, halo, cyano, alkylamino, dialkylamino, trialkylammonium, bis(hydroxyalkyl)amino, and hydroxyalkylaminoalkylamino, wherein each of heteroaryl, heteroaryloxy, and heteroarylamino, heteroarylalkylaminoalkylamino, heterocyclyl, and heterocyclylamino is optionally substituted;
In some other embodiments, the present invention relates to azaindenoisoquinoline compounds having the formula (II), wherein one of R1 is chloro or fluoro and the remaining substituents are hydrogen.
In some other embodiments, the present invention relates to azaindenoisoquinoline compounds having the formula (II), wherein one of R1 is nitro and the remaining substituents are hydrogen.
In some other embodiments, the present invention relates to azaindenoisoquinoline compounds having the formula (II), wherein R2 is a halo or cyano.
In some other embodiments, the present invention related to azaindenoisoquinoline compounds having the formula (II), wherein R2 is a substituent selected from the group consisting of:
Figure US10875860-20201229-C00006
In some other embodiments, the present invention relates to azaindenoisoquinoline compounds having the formula (I), wherein one of R3 is an alkoxy and the remaining R3 substituents are hydrogen.
In some other embodiments, the present invention relates to azaindenoisoquinoline compounds having the formula (II), wherein one of R3 is alkoxy and the remaining R3 substituents are hydrogen.
In some other embodiments, the present invention relates a pharmaceutical composition comprising a compound of formula (I), or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers, diluents, and excipients.
In some other embodiments, the present invention relates a pharmaceutical composition comprising a compound of formula (II), or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers, diluents, and excipients.
In some other embodiments, the present invention relates a method for treating a patient with a cancer, comprising the step of administering a therapeutically effective amount of the pharmaceutical composition disclosed herein to the patient in need of relief from said cancer.
In some other embodiments, the present invention relates a method for treating a patient with a cancer, the method comprising the step of administering a therapeutically effective amount of a compound disclosed herein, together with one or more pharmaceutically acceptable carriers, diluents, and excipients, to the patient in need of relief from said cancer.
In some other embodiments, the present invention relates a method for treating a patient with a cancer, the method comprising the step of administering a therapeutically effective amount of a compound disclosed herein, and a therapeutically effective amount of one or more other compounds of the same or different mode of action, together one or more pharmaceutically acceptable carriers, diluents, and excipients, to the patient in need of relief from said cancer.
In some embodiments, the present invention relates a method for treating a patient with a cancer, the method comprising the step of administering a therapeutically effective amount of a compound functioning as a triple inhibitor toward human topoisomerase 1, tyrosyl-DNA phosphodiesterase 1, and tyrosyl-DNA phosphodiesterase 2.
In some other embodiments, the present invention relates a substance for use in the treatment of a cancer, the substance having formula (I)
Figure US10875860-20201229-C00007

or a pharmaceutically acceptable salt thereof, wherein:
    • m is an integer from 1 to about 6;
  • R1 represents 1-4 substituents each of which is independently selected from the group consisting of hydrogen, halo, hydroxy, nitro, cyano, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkylthio, optionally substituted alkylsulfonyl, carboxylic acid, and sulfonic acid; or R1 represents 2-4 substituents where 2 of said substituents are adjacent substituents and are taken together with the attached carbons to form an optionally substituted heterocycle, and where any remaining substituents are each independently selected from the group consisting of hydrogen, halo, hydroxy, optionally substituted alkyl, optionally substituted alkoxy, cyano, nitro, optionally substituted alkylthio, optionally substituted alkylsulfonyl, carboxylic acid, and sulfonic acid;
  • R2 is selected from the group consisting of heteroaryl, heteroaryloxy, heteroarylamino, heteroarylalkylaminoalkylamino, heterocyclyl, heterocyclylamino, amino, hydroxyl, halo, cyano, alkylamino, dialkylamino, trialkylammonium, bis(hydroxyalkyl)amino, and hydroxyalkylaminoalkylamino, wherein each of heteroaryl, heteroaryloxy, and heteroarylamino, heteroarylalkylaminoalkylamino, heterocyclyl, and heterocyclylamino is optionally substituted; and
  • R3 represents 1-3 substituents each of which is independently selected from the group consisting of hydrogen, halo, hydroxy, cyano, nitro, optionally substituted alkyl, optionally substituted alkoxy, optionally substituted alkylthio, optionally substituted alkylsulfonyl, carboxylic acid, and sulfonic acid; or R3 represents 2-3 substituents where 2 of said substituents are adjacent substituents and are taken together with the attached carbons to form an optionally substituted heterocycle, and where the remaining substituent is independently selected from the group consisting of hydrogen, halo, hydroxy, optionally substituted alkyl, optionally substituted alkoxy, cyano, nitro, optionally substituted alkylthio, optionally substituted alkylsulfonyl, carboxylic acid, and sulfonic acid, and
wherein at least one of R1 or R3 is not hydrogen.
The present invention may be better understood in light of the following non-limiting compound examples and method examples.
Preparation of compound 7 was first explored through Mitsunobu reaction by treating 6 (Kiselev, et al, J. Med. Chem. 2012, 55, 1682-1697) with 3-bromopropanol, Ph3P, and DIAD in THF (Scheme 1), but only a moderate yield (39%) was obtained and tedious chromatography was required to get pure product. An SN2 substitution with 1,3-dibromopropane was then investigated to install the 3-bromopropyl side chain. After thorough optimization of the reaction conditions, it was determined that when compound 6 was treated with sodium hydride in anhydrous DMF, followed by addition of sodium iodide (0.1 equiv) and 1,3-dibromopropane at −10° C. to 0° C. for 30 hours, compound 7 could be obtained in 72% yield. This improvement in yield over the previously reported reaction conditions resulted from the use of NaI (Kiselev, et al, J. Med. Chem. 2014, 57, 1289-1298). The practical preparation of compound 7 on a gram scale made it possible to synthesize 7-azaindenoisoquinoline analogues with maximum side chain variation, thereby facilitating the structure-activity relationship (SAR) study.
Figure US10875860-20201229-C00008
Figure US10875860-20201229-C00009
The SAR study was restricted to varying the lactam γ-aminopropyl side chain. As illustrated in Scheme 2, compounds 8, 9, 10, 12, 13, 16 and 17 were prepared in 50-77% yields by heating compound 7 with the corresponding amines at reflux in 1,4-dioxane in the presence of K2CO3 and NaI, while compounds 11, 14 and 15 were obtained by treatment of 7 with methylamine, isopropylamine, and ethylamine in DMF at room temperature in the presence of triethylamine and 0.1 equivalent of NaI in 68%, 80% and 73% yields, respectively.
The anhydrides 11a and 11b (Scheme 3) were prepared by published literature procedures (Kang, B.-R, et al., Med. Chem. Res. 2014, 23, 1340-1349). Bromination of 5-methoxy-3-methylpicolinonitrile in the presence of the radical initiator AIBN produced intermediate bromide 12a (Kiselev, et al, J. Med. Chem. 2011, 54, 6106-6116), which was used directly in the next step without additional purification. The condensation of 12a and 11a-b in acetonitrile promoted by Et3N afforded compounds 13a-b in ˜25% yield for two steps. Oxidation of 13a-b with selenium dioxide provided azaindenoisoquinoline intermediates 14a-b. Treating compounds 14a-b with NaH in DMF at 0° C. followed by reaction with 1-chloro-3-bromopropane yielded the common intermediates 15a-b. The common intermediates 15a-b were used for the synthesis of the final compounds 16a-j and 17a-i by alkylation of the corresponding amines in DMF as shown in Scheme 4.
Figure US10875860-20201229-C00010
Figure US10875860-20201229-C00011
Biological Results and Discussion
All of the synthesized compounds 7-17 were tested for their inhibitory activities against Top1, Tdp1, and Tdp2. Their antiproliferative activities were also evaluated against the NCI-60 panel of human cancer cell lines. Top1 inhibition was recorded as the ability of the drug to induce enzyme-linked DNA breaks in the Top1-mediated DNA cleavage assay (Dexheimer, et al., Nat. Protoc. 2008, 3, 1736-1750). The results of this assay are designated relative to the Top1 inhibitory activities of camptothecin (18) and MJ-III-65 (19) (also known as NSC 706744 and LMP744, respectively) and are expressed in semi quantitative fashion: 0, no detectable activity; +, weak activity; ++, less activity than that of compound 19; +++, similar activity to that of 19; ++++, equipotent to 18. As shown in Table 1, all of the compounds 7-17 exhibited moderate to potent Top1 inhibitory activity. Compound 7 expressed Top1 inhibitory activity at the ++ potency level. Further evaluation of analogues with various amino groups in the side chain (pyrrolidine, 8; 1-methylpiperazine, 9; and piperazine, 10) revealed similar Top1 inhibitory activities at the ++ level. Interestingly, compound 11, in which a methylamine was introduced at the end of propyl chain, displayed improved Top inhibitory activity relative to 7 at the +++ level. Notably, the isopropylamine 14 and ethylamine 15 compounds demonstrated excellent Top1 inhibitory activity at the ++++ level. The 4-2(aminoethyl)morpholine compound 13 and 2-amino-2-thiazoline compound 12 displayed improved Top1 inhibitory activity relative to 7 at the +++ and ++++ levels, respectively. The improved potencies of compounds 11-15 compared to those of 8-10 indicate that a secondary amine in side chain of 7-azaindenoisoquinolines is better than a tertiary amine for the Top1 inhibitory activity.
The Tdp1 inhibitory activities of the 7-azaindenoisoquinolines were determining by measuring their abilities to inhibit the hydrolysis of the phosphodiester linkage between the tyrosine residue and the 3′-end of a DNA oligonucleotide substrate (N14Y), thus preventing the generation of an oligonucleotide with a free 3′-phosphate (N14P) (Dexheimer, et al., J. Med. Chem. 2009, 52, 7122-7131). The Tdp1 inhibitory activities of the 7-azaindenoisoquinolines are summarized in Table 1. Tdp1 inhibitory potencies were determined in duplicate using a semiquantitative scale: 0, IC50>111 μM; +, IC50 between 37 and 111 μM; ++, IC50 between 12 and 37 μM; +++, IC50 between 1 and 12 μM; and ++++, IC50<1 μM.
TABLE 1
Top1, Tdp1, and Tdp2 inhibitory activities and cytotoxicities of 7-azaindenoisoquinolines (series 1)
Cytotoxicity (GI50, μM)a
lung CNS Melanoma Ovarian Renal Breast Enzyme Inhibitory Activity
Compd HOP-62 SF-539 UACC-62 OVCAR-3 SN12C MCF-7 MGM Top1c Tdp1d Tdp2d
7 0.058 0.279 0.082 0.248 0.269 0.046 0.112 ++ ++ +
8 <0.01 0.031 0.015 0.059 0.036 <0.01 0.045 ++ +++ +
9 0.081 0.191 0.183 0.171 0.214 0.016 0.562 ++ ++ ++
10 0.065 0.355 0.175 1.190 0.230 0.036 0.316 ++ ++ +
11 0.030 0.051 0.020 0.174 0.046 1.06 0.741 +++ +++ +
12 0.014 0.036 <0.01 0.137 <0.01 <0.01 0.058 ++++ +++ ++
13 0.150 0.311 0.044 0.315 0.080 0.032 0.177 +++ ++ +
14 0.085 0.051 0.019 0.082 0.039 0.012 0.058 ++++ ++ +
15 0.107 0.070 0.027 0.062 0.053 0.019 0.079 ++++ +++ +
16 2.76 1.70 1.22 1.46 0.142 1.66 +++ 0 0
17 NSb NS NS NS NS NS NS ++ +++ +++
18 <0.01 <0.01 <0.01 0.22 0.020 0.013 0.040 ++++ 0 0
19 0.02 0.04 0.03 0.50 <0.01 <0.01 0.21 ++++ +/++ +++
aThe cytotoxicity GI50 values listed are the concentrations corresponding to 50% growth inhibition and are the result of single determinations;
bNot selected for 5-concentration testing due to low potency in the initial 1-concentration test.
cCompound-induced DNA cleavage resulting from Top1 inhibition is graded by the following semiquantitative scale relative to 1 μM MJ-III-65 (19) and 1 μM camptothecin (18): 0, no detectable activity; +, weak activity; ++, activity less than that of MJ-III-65 (19); +++, activity equal to that of 19; ++++, activity equipotent with 18.
dTdp1 and Tdp2 IC50 values were determined in duplicate using a semiquantitative scale: 0, IC50 > 111 μM; +, IC50 between 37 and 111 μM; ++, IC50 between 12 and 37 μM; +++, IC50 between 1 and 12 μM; ++++, IC50 < 1 μM.
TABLE 2
Top1, Tdp1, and Tdp2 inhibitory activities and cytotoxicities of 7-azaindenoisoquinolines (series 2)
Cytotoxicity (GI50, μM)a
Lung Colon CNS Melanoma Ovarian Renal Prostate Breast Enzyme Inhibitory Activity
Compd HOP-62 HCT-116 SF-539 UACC-62 Ovcar-3 SN12C DU-145 MCF-7 MGM Top1b Tdp1c Tdp2d
16a 0.27 0.16 0.23 0.18 0.78 NA 0.41 0.09 0.10 +++ >111 >111
17a 0.187 0.115 0.228 0.235 0.250 0.320 0.341 0.044 0.269 +++ >111 >111
16b 0.02 0.014 0.038 0.028 0.12 NA 0.043 <0.01 0.063 +++ >111 >111
17b <0.01 <0.01 0.024 0.016 0.043 <0.01  <0.01 <0.01 0.033 +++ >111 >111
16c 0.042 0.35 0.47 0.095 1.3 NA 0.092 0.036 0.39 ++ 11.9 ± 3.9 32.9 ± 5.7
17c 0.059 0.082 0.305 0.344 0.567 0.238 0.144 0.033 0.269 ++ >111 61.6 ± 14 
16d 0.094 0.098 0.43 0.19 1.42 NA 0.27 0.042 0.66 ++++ 14 ± 1 >111
17d 0.142 0.089 0.295 0.240 0.254 0.189 0.174 0.031 0.199 + >111 >111
16e 0.20 0.298 1.33 1.43 1.43 0.489 0.242 0.089 0.58 + 20.2 ± 3.7 >111
17e 0.066 0.091 0.532 1.26 0.335 0.080 0.062 0.034 0.218 + >111  107 ± 5.2
16f 0.070 0.138 0.554 0.303 1.39 0.417 0.173 0.037 0.380 ++ 33.3 ± 4.2 >111
17f 0.107 0.093 0.248 0.271 0.190 0.164 0.210 0.031 0.165 ++ >111 >111
16g 0.220 0.550 0.730 0.574 1.41 1.18  0.331 0.046 0.60 ++/+++   16 ± 3.2 >111
17g 0.142 0.143 0.368 0.608 0.263 0.211 0.202 0.034 0.245 + >111 108 ± 3 
16h 0.122 0.203 0.506 0.532 2.13 0.675 0.208 <0.01 0.426 ++/+++ 22.2 ± 6.4 >111
17h 0.149 0.136 0.320 0.341 0.282 0.151 0.215 0.042 0.229 ++ >111  104
16i 0.046 0.104 1.98 2.09 5.97 2.05  0.186 0.023 0.630 ++ 27.5 ± 0.6 >111
17i 0.212 0.153 0.360 0.471 0.307 0.241 0.211 0.048 0.288 +++ >111 >111
16j 0.950 0.980 2.15 1.81 15.1 10.2   3.18 0.487 2.95 + >111 >111
aThe cytotoxicity GI50 values listed are the concentrations corresponding to 50% growth inhibition and are the result of single determinations;
bCompound-induced DNA cleavage resulting from Top1 inhibition is graded by the following semiquantitative scale relative to 1 μM MJ-III-65 (19) and 1 μM camptothecin (18): 0, no detectable activity; +, weak activity; ++, activity less than that of MJ-III-65 (19); +++, activity equal to that of 19; ++++, activity equipotent with 18 (see Table 1).
cTdp1 and Tdp2 IC50 values were determined in duplicate.
To investigate their potential as anticancer agents, the 7-azaindenoisoquinolines 7-17 were evaluated in the National Cancer Institute's Developmental Therapeutics Program screen against 60 human cancer cell lines (the “NCI-60,” Shoemaker, R. H. Nat. Rev. Cancer 2006, 6, 813-823). The GI50 values obtained with selected cell lines, along with the mean graph midpoint (MGM) values, are summarized in Table 1. The MGM was based on a calculation of the average GI50 for all of the cell lines tested in which GI50 values below and above the test range (0.01 μM to 100 μM) were taken as the minimum (0.01 μM) and maximum (100 μM) drug concentrations used in the screening test. Many of these new compounds display significant potencies against various cell lines with GI50 MGMs in the low micromolar to submicromolar range (0.045-1.66 μM). The Top1 ++++ compounds 12, 14, and 15 were among the most cytotoxic, with MGM values of 0.058, 0.058, and 0.079 μM, respectively. The differences in cytotoxicity among these three compounds are low despite clear differences in Tdp1 and Tdp2 inhibition, suggesting that Tdp1 and Tdp2 inhibition do not contribute significantly to the cytotoxicities of these compounds. The Top1 +++ compounds are 11 (MGM 0.714 μM), 13 (MGM 0.177 μM), and 16 (MGM 1.650 μM). On the basis of the Tdp1 and Tdp2 inhibitory potencies, 16 is expected to be the least potent, which it clearly is. However, one would expect 11 to be the most cytotoxic, and in fact 13 is the most cytotoxic of the three compounds. Among the Top1++ compounds, 7 (MGM 0.112 M), 8 (MGM 0.045 μM), 9 (MGM 0.562 μM), 10 (MGM 0.316 μM), and 17 (not tested due to low cytotoxicity in the one-concentration preliminary testing), 17 is expected to be the most cytotoxic on the basis of Tdp1 and Tdp2 inhibition, but it is actually the least cytotoxic. Based on their performance against Top1, Tdp1, and Tdp2, compounds 7 or 10 should be the least cytotoxic, but actually 17 is. On the other hand, compound 8 is surprisingly the most cytotoxic compound in the whole series despite having relatively moderate Top1 inhibitory activity. Overall, these triple inhibitors are very cytotoxic anticancer agents, but their relative cytotoxicities cannot be rationalized simply on the basis of their inhibitory activities vs. the three enzymes. Other factors that could contribute to the lack of agreement between the enzyme inhibitory activities and cytotoxicities of these 7-azaindenoisoquinoline include differences in cellular uptake, distribution within the cell, metabolism, efflux from the cell, off-target effects, and lack of sufficient potency vs. Tdp1 and Tdp2 to exert a significant synergistic effect.
Compounds 16a-j and 17a-i were tested in a Top1-mediated DNA cleavage assay to assess Top1 poisoning activity and the results were summarized in Table 2. In addition, those compounds were tested for anti-proliferative activity in the NCI-60 human tumor cell line screen. The Top1-mediated DNA cleavage assay scores the activity of Top poisons with a rubric based on the activity of 1 μM camptothecin. Test agents are incubated at 0.1, 1, 10, and 100 μM concentrations with a 3′-[32P]-labeled double-stranded DNA fragment and Top1 enzyme. Top1 poisons bind to and trap Top1-DNA cleavage complexes. The DNA cleavage pattern is then documented by gel electrophoresis. Visual comparison of the lanes produced with 1 μM CPT indicates the activity of the new compounds. Finally, a semiquantitative score which ranges from 0 (no activity) to ++++(activity equal to that of 1 μM CPT) is used to describe the activity of the new compounds (see Table 2 caption for a complete description of the scoring rubric). The biological activities of the compounds are summarized in Table 2. The compounds have a good to moderate Top1 inhibitory activity. Of the nineteen compounds synthesized, compound 16d displayed the best activity with a score of ++++, while five other compounds exhibited good activity with a score of +++. On the other hand, five compounds showed weak activity with only a “+” score. Some of the compounds act as Top1 suppressors at high concentration, which likely results from the binding of the drug to the DNA at high drug concentration, making the DNA a poorer substrate for the cleavage reaction.
Compounds Examples
General.
NMR spectra were obtained at 300 or 500 (1H) and 75 or 125 (13C) MHz using Bruker ARX300 or Bruker DX-2 500 [QNP probe or multinuclear broadband observe (BBO) probe, respectively] spectrometers. Column chromatography was performed with 230-400 mesh silica gel. The melting points were determined using capillary tubes with a Mel-Temp apparatus and are uncorrected. IR spectra were obtained using a Perkin-Elmer 1600 series FTIR spectrometer on salt plates or as KBr pellets. ESI-MS analyses were recorded on a FinniganMAT LCQ Classic mass spectrometer. APCI-MS analyses were performed using an Agilent 6320 ion trap mass spectrometer. EI/CIMS analyses were obtained with a Hewlett-Packard Engine mass spectrometer. All mass spectral analyses were performed at the Campus-Wide Mass Spectrometry Center of Purdue University. HPLC analyses were carried out on a Waters 1525 binary HPLC pump/Waters 2487 dual λ absorbance detector system using a 5 μm C18 reverse phase column. All reported yields refer to pure isolated compounds. Chemicals and solvents were of reagent grade and used as obtained from commercial sources without further purification. The purities of all of the biologically tested compounds were >95% as estimated by HPLC or determined by elemental analysis. For HPLC, the peak area of the major product was >95% of the combined total peak areas when monitored by a UV detector at 254 nm.
7-Aza-6-(3-bromopropyl)-5,6-dihydro-9-methoxy-3-nitro-5,11-dioxo-11H-indeno[1,2-c]isoquinoline (7)
(Kiselev, et al., J. Med. Chem. 2014, 57, 1289-1298). Sodium hydride (234 mg, 9.28 mmol) and sodium iodide (70 mg, 0.464 mmol) were added to a suspension of 7-aza-5,6-dihydro-9-methoxy-3-nitro-5,11-dioxo-11H-indeno[1,2-c]-isoquinoline (6, 1.5 g, 4.64 mmol) in dry DMF (230 mL) at 0° C. After the reaction mixture had been warmed to room temperature and stirred for 3 h, a dark solution formed. The solution was cooled to −10° C.-0° C. in an ice-salt-water bath, and 1,3-dibromopropane (4.686 g, 23.2 mmol) was added. The solution was stirred for 30 h and the reaction quenched with water (200 mL). The product was extracted with ethyl acetate (5×200 mL). The combined extracts were washed with water (6×150 mL) and brine (3×100 mL), dried with sodium sulfate, and evaporated to dryness under reduced pressure. The residue was triturated with ether, filtered and washed with ether to provide compound 7 as a red solid (1.47 g, 72%): mp 170-172° C. 1H NMR (300 MHz, CDCl3) δ 9.19 (d, J=2.4 Hz, 1H), 8.74 (d, J=8.9 Hz, 1H), 8.48 (dd, J=9.0, 2.3 Hz, 1H), 8.23 (d, J=2.6 Hz, 1H), 7.44 (d, J=2.7 Hz, 1H), 5.16-5.05 (m, 2H), 3.98 (s, 3H), 3.55 (t, J=6.8 Hz, 2H), 2.52-2.37 (m, 2H); ESIMS m/z (rel intensity) 444.0 (MH+, 100).
Mitsunobu Approach.
7-Aza-5,6-dihydro-9-methoxy-3-nitro-5,11-dioxo-11H-indeno[1,2-c]-isoquinoline (6, 97 mg, 0.3 mmol) and triphenylphosphine (236 mg, 0.9 mmol) were diluted in THF (30 mL). 3-Bromopropanol (129 mg, 0.9 mmol) was added, followed by DIAD (182 mg, 0.9 mmol). The solution was stirred at room temperature for 60 h and the reaction mixture was concentrated to dryness. The solid was purified by flash column chromatography, eluting with hexane-ethyl acetate (2:1) to provide the product 7 a red solid (50 mg, 39%).
7-Aza-5,6-dihydro-6-[3-(4-pyrrolidine-1-yl)propyl]-9-methoxy-3-nitro-5,11-dioxo-11H-indeno[1,2-c]isoquinoline (8)
7-Aza-6-(3-bromopropyl)-5,6-dihydro-9-methoxy-3-nitro-5,11-dioxo-11H-indeno[1,2-c]isoquinoline (7, 133 mg, 0.3 mmol), pyrrolidine (214 mg, 3 mmol), NaI (10 mg, 0.03 mmol), and potassium carbonate (208 mg, 1.5 mmol) were diluted with 1,4-dioxane (20 mL). The resulting mixture was heated at reflux for 6 h. The solvent was evaporated under reduced pressure, and the residue was re-dissolved in chloroform (100 mL). The chloroform solution was washed with water (3×10 mL) and brine (20 mL), dried with sodium sulfate, and evaporated to dryness. The solid residue was subjected to column chromatography (silica gel), eluting with 5-20% methanol in dichloromethane to yield the red solid product 8 (88 mg, 68%): mp 248-250° C. IR (film) 3412, 1613, 1483, 1335, 1300 cm−1; 1H NMR (300 MHz, CDCl3) δ 9.17 (d, J=2.1 Hz, 1H), 8.78 (d, J=8.7 Hz, 1H), 8.52 (dd, J=9.0, 2.7 Hz, 1H), 8.27 (d, J=2.6 Hz, 1H), 7.45 (d, J=2.7 Hz, 1H), 5.07 (t, J=6.9 Hz, 2H), 3.99 (s, 3H), 3.87-3.83 (m, 2H), 3.28-3.23 (m, 2H), 2.85-2.80 (m, 2H), 2.55-2.49 (m, 2H), 2.28-2.23 (m, 2H), 2.10-2.07 (m, 2H); ESIMS m/z (rel intensity) 435.1 (MH+, 100). HPLC purity: 99.42% (C18 reverse phase, MeOH—H2O, 85:15); HRMS-ESI m/z: MH+ calcd for C23H23N4O5, 435.1669; found, 435.1656. HPLC purity: 99.42% (C18 reverse phase, MeOH—H2O, 85:15).
Mitsunobu Approach.
7-Aza-5,6-dihydro-9-methoxy-3-nitro-5,11-dioxo-11H-indeno[1,2-c]-isoquinoline (6, 97 mg, 0.3 mmol) and triphenylphosphine (236 mg, 0.9 mmol) were diluted in THF (30 mL). 3-(1-Pyrrolidinyl)-1-propanol (117 mg, 0.9 mmol) was added, followed by DIAD (182 mg, 0.9 mmol). The solution was stirred at room temperature for 72 h. The reaction mixture was concentrated to dryness. The solid was purified by flash column chromatography, eluting with 5-20% methanol in dichloromethane to yield the red solid product 8 (52 mg, 40%).
3-(1-Pyrrolidinyl)-1-propanol
Potassium carbonate (1.4 g, 0.94 mol) and pyrrolidine (0.87 mL, 1.8 mol) were added to a stirred solution of 3-bromopropanol (1 g, 0.635 mol) in 30 mL of THF at 0° C., and the resulting mixture was stirred at room temperature for 15 h. The resulting mixture was diluted with ethyl acetate (200 mL) and filtered through celite. The filtrate was concentrated, and the residue was subjected to column chromatography (silica gel), eluting with 50% methanol in dichloromethane, to yield a yellow oil (0.57 g, 70%). 1H NMR (300 MHz, CDCl3) δ 3.79 (t, J=5.1 Hz, 2H), 2.71 (t, J=5.6 Hz, 2H), 2.57-2.51 (m, 4H), 1.75-1.66 (m, 6H); 13C NMR (125 MHz, CDCl3) δ 64.18, 56.02, 54.09, 29.24, 23.27.
7-Aza-5,6-dihydro-6-[3-(3H-methylpiperazine-1-yl)propyl]-9-methoxy-3-nitro-5,11-dioxo-11H-indeno[1,2-c]isoquinoline (9)
Compound 7 (133 mg, 0.3 mmol), 1-methylpiperazine (300 mg, 3 mmol), NaI (10 mg, 0.03 mmol), and potassium carbonate (208 mg, 1.5 mmol) were diluted with 1,4-dioxane (20 mL). The resulting mixture was heated at reflux for 6 h. The solvent was evaporated under reduced pressure, and the residue was re-dissolved in chloroform (100 mL). The chloroform solution was washed with water (3×10 mL) and brine (20 mL), dried with sodium sulfate, and evaporated to dryness. The solid residue was subjected to column chromatography (silica gel), eluting with 5-20% methanol in dichloromethane, to yield the orange solid product (107 mg, 77%): mp 211-213° C. IR (film) 3429, 1666, 1500, 1334, 1286 cm−1; 1H NMR (300 MHz, CDCl3) δ 9.20 (d, J=2.3 Hz, 1H), 8.77 (d, J=8.9 Hz, 1H), 8.49 (dd, J=8.7, 2.6 Hz, 1H), 8.22 (d, J=2.6 Hz, 1H), 7.46 (d, J=2.7 Hz, 1H), 5.07 (t, J=6.9 Hz, 2H), 3.98 (s, 3H), 2.60-2.55 (m, 2H), 2.46-2.28 (m, 8H), 2.26 (s, 3H), 2.06-2.01 (m, 2H); ESIMS m/z (rel intensity) 464.1 (MH+, 100); HRMS-ESI m/z: MH+ calcd for C24H26N5O5, 464.1934; found, 464.1928. HPLC purity: 98.34% (C18 reverse phase, MeOH—H2O, 90:10).
7-Aza-5,6-dihydro-9-methoxy-3-nitro-5,11-dioxo-6-[3-(piperazine-1-yl)propyl]-11H-indeno[1,2-c]isoquinoline (10)
Compound 7 (133 mg, 0.3 mmol), piperazine (258 mg, 3 mmol), NaI (10 mg, 0.03 mmol) and potassium carbonate (208 mg, 1.5 mmol) were diluted with 1,4-dioxane (20 mL). The resulting mixture was heated at reflux for 6 h. The solvent was evaporated under reduced pressure, and the residue was re-dissolved in chloroform (100 mL). The chloroform solution was washed with water (3×10 mL) and brine (20 mL), dried with sodium sulfate, and evaporated to dryness. The solid residue was subjected to column chromatography (silica gel), eluting with 5-20% methanol in dichloromethane, to yield the red solid product (88 mg, 66%): mp 134-135° C. IR (film) 3418, 2925, 1671, 1613, 1484, 1336 cm−1; 1H NMR (300 MHz, CDCl3) δ 9.18 (d, J=2.4 Hz, 1H), 8.78 (d, J=8.8 Hz, 1H), 8.46 (dd, J=8.9, 2.6 Hz, 1H), 8.20 (d, J=2.5 Hz, 1H), 7.44 (d, J=2.8 Hz, 1H), 5.06 (t, J=6.8 Hz, 2H), 3.95 (s, 3H), 2.58-2.54 (m, 2H), 2.43-2.25 (m, 8H), 2.04-1.98 (m, 2H); 1.91 (brs, 1H); ESIMS m/z (rel intensity) 450.1 (MH+, 100); HRMS-ESI m/z: MH+ calcd for C23H23N5O5, 450.1778; found, 450.1779. HPLC purity: 100% (C18 reverse phase, MeOH—H2O, 80:20).
7-Aza-5,6-dihydro-9-methoxy-6-[3-(methylaminopropyl]-3-nitro-5,11-dioxo-11H-indeno[1,2-c]isoquinoline (11)
Compound 7 (133 mg, 0.3 mmol), NaI (10 mg, 0.03 mmol), methylamine (2M in THF) (3 mL, 6 mmol), and triethylamine (0.125 mL, 0.9 mmol) were diluted with anhydrous DMF (40 mL). The resulting mixture was stirred for 12 h at room temperature and the reaction quenched with water (5 mL). The products were extracted with chloroform (5×40 mL). The combined extracts were washed with water (6×20 mL) and brine (3×20 mL), dried with sodium sulfate, and evaporated to dryness under reduced pressure. The solid residue was subjected to column chromatography (silica gel), eluting with 5-20% methanol in dichloromethane containing 1% trimethylamine, to yield a red solid product (80 mg, 68%): mp 259-262° C. IR (film) 3435, 2964, 2782, 1682, 1555, 1481, 1336, 1302 cm1; 1H NMR (300 MHz, CDCl3) δ 9.18 (d, J=2.4 Hz, 1H), 8.75 (d, J=8.9 Hz, 1H), 8.48 (dd, J=8.70, 2.7 Hz, 1H), 8.23 (d, J=2.7 Hz, 1H), 7.43 (d, J=2.4 Hz, 1H), 5.03 (t, J=7.5 Hz, 2H), 3.97 (s, 3H), 2.77-2.72 (m, 2H), 2.45 (s, 3H), 2.09-1.96 (m, 3H); ESIMS m/z (rel intensity) 395.1 (MH+, 100); HRMS-ESI m/z: MH+ calcd for C20H18N4O5, 395.1356; found, 395.1357. HPLC purity: 96.98% (C18 reverse phase, MeOH—H2O, 85:15).
7-Aza-5,6-dihydro-6-[3-((4,5-dihydrothiazol-2-yl)amino)propyl]-9-methoxy-3-nitro-5,11-dioxo-11H-indeno[1,2-c]isoquinoline (12)
Compound 7 (106 mg, 0.24 mmol), 2-amino-2-thiazoline (333 mg, 2.4 mmol), NaI (9 mg, 0.024 mmol), and potassium carbonate (165 mg, 1.2 mmol) were diluted with 1,4-dioxane (30 mL). The resulting mixture was heated at reflux for 6 h. The solvent was evaporated under reduced pressure, and the residue was re-dissolved in chloroform (100 mL). The chloroform solution was washed with water (3×10 mL) and brine (20 mL), dried with sodium sulfate, and evaporated to dryness. The solid residue was subjected to column chromatography (silica gel), eluting with 5-20% methanol in dichloromethane, to yield the red solid product (63 mg, 57%): mp 175-177° C. IR (film) 3430, 1641, 1613, 1504, 1482, 1335 cm1; 1H NMR (300 MHz, CDCl3) δ 9.19 (d, J=2.2 Hz, 1H), 8.75 (d, J=8.9 Hz, 1H), 8.49 (dd, J=8.9, 2.1 Hz, 1H), 8.28 (d, J=2.7 Hz, 1H), 7.43 (d, J=2.8 Hz, 1H), 5.05 (t, J=5.2 Hz, 2H), 3.97 (s, 3H), 3.76 (t, J=6.7 Hz, 2H), 3.64 (t, J=6.5 Hz, 2H), 3.42 (t, J=7.5 Hz, 1H), 3.24 (t, J=6.7 Hz, 2H), 2.20-2.15 (m, 2H); ESIMS m/z (rel intensity) 466.1 (MH+, 100); HRMS-ESI m/z: MH+ calcd for C22H19N5O5S, 466.1185; found, 466.1182. HPLC purity: 97.50% (C18 reverse phase, MeOH—H2O, 90:10).
7-Aza-5,6-dihydro-9-methoxy-6-[3-((2-morpholinoethyl)amino)propyl]-3-nitro-5,11-dioxo-11H-indeno[1,2-c]isoquinoline (13)
Compound 7 (106 mg, 0.24 mmol), 4-(2-aminoethyl)-morpholine (314 mg, 2.4 mmol), NaI (9 mg, 0.024 mmol), and potassium carbonate (165 mg, 1.2 mmol) were diluted with 1,4-dioxane (30 mL). The resulting mixture was heated at reflux for 6 h. The solvent was evaporated under reduced pressure, and the residue was re-dissolved in chloroform (100 mL). The chloroform solution was washed with water (3×10 mL) and brine (20 mL), dried with sodium sulfate, and evaporated to dryness. The solid residue was subjected to column chromatography (silica gel), eluting with 5-20% methanol in dichloromethane, to yield the red solid product (58 mg, 50%): mp 214-216° C. IR (film) 2770, 1437, 1332, 1219 cm−1; 1H NMR (300 MHz, CDCl3) δ 9.04 (d, J=2.2 Hz, 1H), 8.80 (d, J=8.9 Hz, 1H), 8.51 (dd, J=8.9, 2.1 Hz, 1H), 8.26 (d, J=2.7 Hz, 1H), 7.47 (d, J=2.8 Hz, 1H), 4.97 (t, J=5.3 Hz, 2H), 3.99 (s, 3H), 3.94-3.89 (m, 4H), 3.78 (t, J=6.5 Hz, 2H), 3.20-3.15 (m, 2H), 3.08-3.02 (m, 2H), 2.80-2.75 (m, 3H), 2.65 (m, 2H), 2.22-2.18 (m, 2H); ESIMS m/z (rel intensity) 494.2 (MH+, 100); HRMS-ESI m/z: MH+ calcd for C25H27N5O6, 494.2040; found, 494.2038. HPLC purity: 98.44% (C18 reverse phase, MeOH—H2O, 90:10).
7-Aza-5,6-dihydro-6-[3-(isopropylamino)propyl]-9-methoxy-3-nitro-5,11-dioxo-11H-indeno[1,2-c]isoquinoline (14)
Compound 7 (133 mg, 0.3 mmol), NaI (10 mg, 0.03 mmol), isopropylamine (177 mg, 6 mmol), and triethylamine (0.125 mL, 0.9 mmol) were diluted with anhydrous DMF (40 mL). The resulting mixture was stirred for 12 h at room temperature and the reaction quenched with water (5 mL). The products were extracted with chloroform (5×40 mL). The combined extracts were washed with water (6×20 mL) and brine (3×20 mL), dried with sodium sulfate, and evaporated to dryness under reduced pressure. The solid residue was subjected to column chromatography (silica gel), eluting with 5-20% methanol in dichloromethane containing 1% trimethylamine, to yield the red solid product (100 mg, 80%): mp 278-280° C. IR (film) 2961, 2805, 1677, 1610, 1337, 1288, 854 cm1; 1H NMR (300 MHz, DMSO) δ 8.90 (d, J=2.1 Hz, 1H), 8.67 (d, J=8.9 Hz, 1H), 8.61 (dd, J=8.8, 2.3 Hz, 1H), 8.37 (d, J=2.4 Hz, 1H), 7.70 (d, J=2.7 Hz, 1H), 4.89 (t, J=7.5 Hz, 2H), 3.98 (s, 3H), 3.28-3.21 (m, 1H), 3.09-3.01 (m, 2H), 2.16-2.10 (m, 2H), 1.20 (d, J=6.5 Hz, 6H); ESIMS m/z (rel intensity) 423.2 (MH+, 100); HRMS-ESI m/z: MH+ calcd for C22H22N4O5, 423.1669; found, 423.1668. HPLC purity: 98.73% (C18 reverse phase, MeOH—H2O, 80:20).
7-Aza-6-[3-(ethylamino)propyl]-5,6-dihydro-9-methoxy-3-nitro-5,11-dioxo-11H-indeno[1,2-c]isoquinoline (15)
A mixture of compound 7 (133 mg, 0.3 mmol), anhydrous DMF (35 mL), NaI (10 mg, 0.03 mmol), ethylamine (3 mL of a 2 M solution in THF, 6 mmol), and triethylamine (0.125 mL, 0.9 mmol) was stirred for 12 h at room temperature and the reaction was then quenched with water (5 mL). The product was extracted with chloroform (6×40 mL). The combined extracts were washed with water (5×30 mL) and brine (3×20 mL), dried with sodium sulfate, and evaporated to dryness under reduced pressure. The solid residue was subjected to column chromatography (silica gel), eluting with 5-20% methanol in dichloromethane containing 1% trimethylamine, to yield the red solid product (89 mg, 73%): mp 266-268° C. IR (film) 3583, 2946, 1682, 1571, 1482, 1336, 1300 cm−1; 1H NMR (300 MHz, DMSO) δ 8.89 (d, J=2.1 Hz, 1H), 8.65-8.59 (m, 2H), 8.36 (d, J=2.6 Hz, 1H), 7.69 (d, J=2.7 Hz, 1H), 4.89 (t, J=7.5 Hz, 2H), 3.98 (s, 3H), 3.08-3.01 (m, 2H), 2.97-2.89 (m, 2H), 2.15-2.09 (m, 2H), 1.17 (t, J=6.5 Hz, 3H); MALDI: m/z 409 (MH+); HRMS-ESI m/z: MH+ calcd for C21H20N4O5, 409.1512; found, 409.1516. HPLC purity: 98.32% (C18 reverse phase, MeOH—H2O, 90:10).
7-Aza-5,6-dihydro-6-[3-(4-(2-hydroxyethyl)piperazin-1-yl)propyl]-9-methoxy-3-nitro-5,11-dioxo-11H-indeno[1,2-c]isoquinoline (16)
Compound 7 (133 mg, 0.3 mmol), 1-(2-hydroxyethyl)piperazine (270 mg, 6 mmol), NaI (9 mg, 0.024 mmol), and potassium carbonate (165 mg, 1.2 mmol) were diluted with 1,4-dioxane (30 mL). The resulting mixture was heated at reflux for 6 h. The solvent was evaporated under reduced pressure, and the residue was re-dissolved in chloroform (120 mL). The chloroform solution was washed with water (3×10 mL) and brine (20 mL), dried with sodium sulfate, and evaporated to dryness. The solid residue was subjected to column chromatography (silica gel), eluting with 5-20% methanol in dichloromethane to yield the red solid product (78 mg, 53%): mp 214-216° C. IR (film) 3439, 1636, 1507, 1483, 1256 cm−1; 1H NMR (300 MHz, CDCl3) δ 9.20 (d, J=2.3 Hz, 1H), 8.76 (d, J=8.9 Hz, 1H), 8.50 (dd, J=8.7, 2.7 Hz, 1H), 8.21 (d, J=2.8 Hz, 1H), 7.44 (d, J=2.7 Hz, 1H), 4.90 (t, J=7.5 Hz, 2H), 3.98 (s, 3H), 3.59-3.55 (m, 2H), 2.97 (t, J=2.5 Hz, 1H), 2.59-2.44 (m, 12H), 2.08-2.03 (m, 2H); ESIMS m/z (rel intensity) 494.2 (MH+, 100); HRMS-ESI m/z: MH+ calcd for C25H27N5O6, 494.2040; found, 494.2041. HPLC purity: 96.97% (C18 reverse phase, MeOH—H2O, 90:10).
6-[3-(4-Aminopiperidin-1-yl)propyl]-7-aza-5,6-dihydro-9-methoxy-3-nitro-5,11-dioxo-11H-indeno[1,2-c]isoquinoline (17)
Compound 7 (133 mg, 0.3 mmol), 4-aminopiperidine (600 mg, 6 mmol), NaI (9 mg, 0.024 mmol), and potassium carbonate (165 mg, 1.2 mmol) were diluted with 1,4-dioxane (30 mL). The resulting mixture was heated at reflux for 6 h. The solvent was evaporated under reduced pressure, and the residue was re-dissolved in chloroform (140 mL). The chloroform solution was washed with water (3×10 mL) and brine (20 mL), dried with sodium sulfate, and evaporated to dryness. The solid residue was subjected to column chromatography (silica gel), eluting with 5-15% methanol in dichloromethane containing 1% trimethylamine to yield the red solid product (87 mg, 63%): mp 258-260° C. IR (film) 3440, 1641, 1436, 1335, 1301, 1084 cm−1; 1H NMR (300 MHz, DMSO) δ 8.87 (d, J=2.2 Hz, 1H), 8.65-8.57 (m, 3H), 8.36 (d, J=2.6 Hz, 1H), 8.24 (d, J=2.3 Hz, 1H), 7.67 (d, J=2.6 Hz, 1H), 4.87 (t, J=7.4 Hz, 2H), 3.98 (s, 3H), 2.50-2.39 (m, 7H), 2.04-1.90 (m, 6H); ESIMS m/z (rel intensity) 464.1; HRMS-ESI m/z: MH+ calcd for C24H26N5O5, 464.1934; found, 464.1919. HPLC purity: 95.10% (C18 reverse phase, MeOH—H2O, 80:20).
7-Aza-3-chloro-5,6-dihydro-9-methoxy-5-oxo-1H-indeno-[1,2-c]isoquinoline (13a)
5-Methoxy-3-methylpicolinonitrile (1.06 g, 7.2 mmol), NBS (1.41 g, 7.92 mmol), and AIBN (118 mg, 0.72 mmol) were diluted with 1,2-dichloroethane (40 mL) and the mixture was heated at reflux for 24 h. The reaction mixture was filtered and the filtrate was evaporated to dryness under reduced pressure to give crude 12a. The residue was re-dissolved in acetonitrile (70 mL). 7-Chloroisochroman-1,3-dione (11a, 2.12 g, 10.8 mmol) was added, followed by triethylamine (1.2 mL, 9.36 mmol), and the solution was heated at reflux for 24 h. The hot solution was filtered and the precipitate was washed with boiling acetonitrile (3×40 mL) to provide a gray solid 13a (698 mg, 32%): mp 258-262° C. The product was introduced into the next step without additional purification. APCIMS m/z (rel intensity): 299 (MH+, 100).
7-Aza-3-fluoro-5,6-dihydro-9-methoxy-5-oxo
11H-indeno-[1,2-c]isoquinoline (13b). 5-Methoxy-3-methylpicolinonitrile (2.09 g, 14.12 mmol), NBS (2.6 g, 14.60 mmol), and AIBN (214 mg, 1.31 mmol) were diluted with CCl4 (100 mL) and the mixture was heated at reflux for 24 h. The reaction mixture was filtered and the filtrate was evaporated to dryness under reduced pressure to give crude 12a. The residue was re-dissolved in acetonitrile (140 mL). 7-Fluoroisochroman-1,3-dione (11b, 2.8 g, 14.26 mmol) was added, followed by triethylamine (1.6 mL, 12.48 mL), and the solution was heated at reflux for 24 h. The hot solution was filtered and the precipitate was washed with boiling acetonitrile (3×60 mL) to provide a gray solid 13b (1.26 g, 31.6%): mp 304-310° C. The product was introduced into the next step without additional purification. 1H NMR (300 MHz, DMSO) δ 12.31 (s, 1H), 8.43-8.17 (m, 2H), 7.91 (ddd, J=14.1, 9.2, 4.0 Hz, 3H), 7.80-7.55 (m, 2H), 3.93 (s, 3H); APCIMS m/z (rel intensity) 283.0 (MH+, 100).
7-Aza-3-chloro-5,6-dihydro-9-methoxy-5,11-dioxo-11H-indeno[1,2-c]isoquinoline (14a)
7-Aza-3-chloro-5,6-dihydro-9-methoxy-5-oxo-11H-indeno[1,2-c]isoquinoline (13a, 698 mg, 2.34 mmol) and SeO2 (525 mg, 4.68 mmol) were diluted with 1,4-dioxane (40 mL) and the mixture was heated at reflux for 24 h. The reaction mixture was filtered while hot and the precipitate was washed with hot dioxane (4×30 mL). The combined filtrates were evaporated to dryness under reduced pressure to yield 14a (628 mg, 86%) as an orange solid: mp>300° C. IR (thin film) 3434, 1650, 1481, 1307, 1017 cm1; 1H NMR (300 MHz, DMSO) δ 8.38 (d, J=9.1 Hz, 1H), 8.25 (d, J=2.4 Hz, 1H), 8.10 (d, J=2.3 Hz, 1H), 7.87 (dd, J=9.3, 2.6 Hz, 1H), 7.54 (d, J=2.7 Hz, 1H), 3.92 (s, 3H); APCIMS m/z (rel intensity): 311.3 (MH+, 100).
7-Aza-3-fluoro-5,6-dihydro-9-methoxy-5,11-dioxo-11H-indeno[1,2-c]isoquinoline (14b)
7-Aza-3-fluoro-5,6-dihydro-9-methoxy-5-oxo-11H-indeno-[1,2-c]isoquinoline (13b, 1.26 g, 4.47 mmol) and SeO2 (0.94 g, 8.38 mmol) were diluted with 1,4-dioxane (170 mL) and the mixture was heated at reflux for 24 h. The reaction mixture was filtered while hot and the precipitate was washed with hot dioxane (3×50 mL). The combined filtrate was evaporated to dryness under reduced pressure to yield 14b as an orange solid (0.81 g, 40.5%): mp 331-334° C. 1H NMR (300 MHz, DMSO) δ 8.41 (dd, J=8.8, 5.4 Hz, 1H), 8.24 (d, J=2.6 Hz, 1H), 7.85 (dd, J=9.5, 2.8 Hz, 1H), 7.72 (dd, J=10.2, 7.4 Hz, 1H), 7.52 (d, J=2.7 Hz, 1H), 3.93 (s, 3H); IR (thin film) 2994, 1682, 1612, 1574, 1560, 1537 cm1; ESIMS m/z (rel intensity) 295.0 (M-H, 100).
3-Chloro-6-(3-chloropropyl)-9-methoxy-5H-pyrido[3′,2′:4,5]cyclopenta[1,2-c]isoquinoline-5,11(6H)-dione (15a)
Sodium hydride (62 mg, 2.5 mmol) and sodium iodide (24 mg, 0.162 mmol) were added to a suspension of 7-aza-3-chloro-5,6-dihydro-9-methoxy-5,11-dioxo-11H-indeno[1,2-c]-isoquinoline (14a, 270 mg, 0.86 mmol) in dry DMF (30 mL) at 0° C. After the reaction mixture had been warmed to room temperature and stirred for 1.5 h, a dark-red solution formed. The solution was cooled to 0° C. again, and 1-bromo-3-chloropropane (0.4 g, 2.9 mmol) was added. The solution was stirred for 24 h and the reaction was quenched with water (100 mL), followed by extraction with chloroform (3×50 mL). The combined extracts were washed with water (3×50 mL) and brine (50 mL), dried with sodium sulfate, and evaporated to dryness under reduced pressure. The residue was triturated with diethyl ether to yield a red solid product (0.09 g, 26%): mp 195-197° C. IR (thin film) 3062, 1659, 1607, 1483 cm−1; 1H NMR (300 MHz, CDCl3) δ 8.25 (d, J=8.6 Hz, 1H), 8.01 (d, J=2.1 Hz, 1H), 7.84 (d, J=2.7 Hz, 1H), 7.36 (dd, J=8.7, 2.2 Hz, 1H), 7.09 (d, J=2.8 Hz, 1H), 4.82-4.67 (m, 2H), 3.65 (s, 3H), 3.24 (t, J=7.0 Hz, 2H), 2.18-2.03 (m, 2H); MALDIMS m/z (rel intensity) 389/391 (MH+, 100); HRESIMS calcd for C19H15Cl2N2O3 (MH+) 389.0460, found 389.0452.
6-(3-Chloropropyl)-3-fluoro-9-methoxy-5H-pyrido[3′,2′:4,5]cyclopenta[1,2-c]isoquino-line-5,11(6H)-dione (15b)
Sodium hydride (90 mg, 3.56 mmol) and sodium iodide (26 mg, 0.18 mmol) were added to a suspension of 7-aza-3-fluoro-5,6-dihydro-9-methoxy-5,11-dioxo-11H-indeno[1,2-c] isoquinoline (14b, 0.536 g, 1.81 mmol) in dry DMF (45 mL) at 0° C. After the reaction mixture had been warmed to room temperature and stirred for 2 h, a dark-red solution was formed. The solution was cooled to 0° C. again, and 1-bromo-3-chloropropane (726 mg, 2.32 mmol) was added. The solution was stirred for 24 h and quenched with water (150 mL), followed by extraction with ethyl acetate (3×70 mL) and brine (1×70 mL), dried with sodium sulfate, and evaporated to dryness under reduced pressure. The residue was subjected to column chromotography (silica gel), eluting with hexane-ethyl acetate (2:1), to yield the red solid product (0.31 g, 46%): mp 244-248° C. IR (thin film) 3053, 1701, 1653, 1589, 1549, 1507, 1482, 1435, 1285 cm−1; 1H NMR (300 MHz, DMSO) δ 8.50 (dd, J=8.9, 5.4 Hz, 1H), 8.26 (d, J=2.7 Hz, 1H), 7.87 (dd, J=9.5, 2.7 Hz, 1H), 7.73 (td, J=8.8, 2.9 Hz, 1H), 7.52 (d, J=2.8 Hz, 1H), 4.91 (t, J=7.1 Hz, 2H), 3.94 (s, 3H), 3.76 (t, J=6.7 Hz, 2H), 2.28-2.15 (m, 2H); MALDIMS m/z (rel intensity) 373.0 (MH+, 100); HRESIMS calcd for C19H15ClFN2O3(MH+) 373.0755, found 373.0748.
General Procedures for the Preparation of Compounds 16a-j.
Compound 15a (0.1 mmol), K2CO3 (0.05 g, 1 mmol), NaI (40 mg) and the appropriate amine (10 eq) were dissolved in DMF (5 mL). The mixture was stirred overnight at 90° C. and then cooled to room temperature. Water (10 mL) was added to the reaction mixture and then the mixture was extracted with EtOAc (3×10 mL). The combined organic layers were washed with water (3×15 mL) and brine (15 mL), dried over Na2SO4, and evaporated under vacuum. The residue was purified using silica gel column chromatography (MeOH—CHCl3, 5:95).
3-Chloro-9-methoxy-6-(3-morpholinopropyl)-5H-pyrido[3′,2′:4,5]cyclopenta[1,2-c]isoquinoline-5,11(6H)-dione (16a)
This compound was isolated as a red powder (0.03 g, 66%): mp 253-256° C. IR (thin film) 3065, 2941, 2808, 1698, 1661, 1608, 1590, 1562, 1537, 1500 cm−1; 1H NMR (300 MHz, DMSO-d6) δ 8.47 (d, J=8.7 Hz, 1H), 8.26 (d, J=2.8 Hz, 1H), 8.13 (d, J=2.3 Hz, 1H), 7.88 (dd, J=8.7, 2.3 Hz, 1H), 7.59 (d, J=2.8 Hz, 1H), 4.83 (m, 2H), 3.94 (s, 3H), 3.89 (d, J=6.2 Hz, 2H), 3.73-3.56 (m, 3H), 3.20 (m, 2H), 3.03 (m, 3H), 2.20 (m, 2H); MALDIMS m/z (rel intensity) 440 (MH+, 100); HRESIMS calcd for C23H22ClN3O4(MH+) 440.1322, found 440.1372; HPLC purity, 96.2% (MeOH—H2O, 85:15).
6-(3-(1H-Imidazol-1-yl)propyl)-3-chloro-9-methoxy-5H-pyrido[3′,2′:4,5]cyclopenta[1,2-c]isoquinoline-5,11(6H)-dione (16b)
This compound was isolated as an orange-red solid (0.018 g, 41%): mp 284-286° C. IR (thin film) 2918, 1696, 1673, 1607, 1566, 1535, 1501 cm−1; 1H NMR (300 MHz, DMSO-d6) δ 8.37 (d, J=8.7 Hz, 1H), 8.08 (s, 1H), 8.04 (d, J=2.2 Hz, 1H), 7.78 (d, J=6.3 Hz, 1H), 7.62-7.48 (m, 1H), 7.46 (d, J=2.7 Hz, 1H), 7.17-7.01 (m, 1H), 6.85-6.70 (m, 1H), 4.71 (m, 2H), 4.02 (m, 2H), 2.10 (m, 2H); MALDIMS m/z (rel intensity) 421 (MH+, 100); HRESIMS calcd for C22H17ClN4O3(MH+) 421.1068, found 421.1059; HPLC purity, 97.17% (MeOH—H2O, 85:15).
3-Chloro-6-(3-((4,5-dihydrothiazol-2-yl)amino)propyl)-9-methoxy-5H-pyrido[3′,2′:4,5]cyclopenta[1,2-c]isoquinoline-5,11(6H)-dione (16c)
This compound was isolated as a red solid (0.012 g, 25%): mp 295-298° C. IR (thin film) 2924, 1666, 1605, 1536, 1501 cm−1; 1H NMR (300 MHz, DMSO-d6) δ 8.50 (d, J=8.5 Hz, 1H), 8.26 (d, J=2.8 Hz, 1H), 8.15 (d, J=2.0 Hz, 1H), 7.90 (d, J=8.7 Hz, 1H), 7.62 (d, J=2.8 Hz, 1H), 4.85 (m, 2H), 4.06 (m, 2H), 3.96 (s, 3H), 3.88-3.78 (m, 1H), 3.70-3.62 (m, 2H), 3.55-3.38 (m, 2H), 2.09 (m, 2H). MALDIMS m/z (rel intensity) 457 (MH+, 100), 455; HRESIMS calcd for C22H19ClN4O3S (MH+) 455.0945, found 455.0928; HPLC purity, 100% (MeOH—H2O, 85:15).
3-Chloro-6-(3-(4-hydroxypiperidin-1-yl)propyl)-9-methoxy-5H-pyrido[3′,2′:4,5]cyclopenta-[1,2-c]isoquinoline-5,11(6H)-dione (16d)
This compound was isolated as deep red solid (0.18 g, 38%): mp 279-281° C. IR (thin film) 3320, 2940, 1698, 1666, 1610, 1589, 1563, 1538, 1500 cm−1; 1H NMR (300 MHz, DMSO-d6) δ 8.44 (d, J=8.7 Hz, 1H), 8.24 (d, J=2.8 Hz, 1H), 8.11 (d, J=2.3 Hz, 1H), 7.84 (dd, J=8.7, 2.3 Hz, 1H), 7.53 (d, J=2.7 Hz, 1H), 4.87 (m, 2H), 4.58-4.49 (m, 1H), 3.92 (s, 3H), 2.60-2.58 (m, 2H), 1.90-1.85 (m, 2H), 1.49-1.42 (m, 2H), 1.16-1.00 (m, 2H); MALDIMS m/z (rel intensity) 454 (MH+, 100); HRESIMS calcd for C24H24ClN3O4(MH+) 454.1534, found 454.1524; HPLC purity, 96.77% (MeOH—H2O, 85:15).
3-Chloro-9-methoxy-6-(3-(piperazin-1-yl)propyl)-5H-pyrido[3′,2′:4,5]cyclopenta[1,2-c]isoquinoline-5,11(6H)-dione (16e)
This compound was isolated as a red powder (0.044 g, 78%): mp 264-268° C. IR (thin film) 2937, 2808, 2469, 1697, 1669, 1606, 1565, 1536, 1502 cm−1; 1H NMR (300 MHz, DMSO-d6) δ 8.46 (d, J=8.7 Hz, 1H), 8.25 (d, J=2.7 Hz, 1H), 8.12 (d, J=2.3 Hz, 1H), 7.86 (dd, J=8.7, 2.3 Hz, 1H), 7.56 (d, J=2.8 Hz, 1H), 4.87 (m, 2H), 4.10 (m, 2H), 3.94 (s, 3H), 2.76 (m, 4H), 2.35 (m, 4H), 1.89 (m, 2H); MALDIMS m/z (rel intensity) 439 (MH+, 100); HRESIMS calcd for C23H23ClN4O3(MH+) 439.1537, found 439.1525; HPLC purity, 98.56% (MeOH—H2O, 85:15).
3-Chloro-9-methoxy-6-(3-(isopropylamino)propyl)-5H-pyrido[3′,2′:4,5]cyclopenta[1,2-c]isoquinoline-5,11(6H)-dione (16f)
The reaction to prepare this compound was performed in a 45 mL pressure vessel and the product was isolated as red powder (0.032 g, 60%): mp 250-251° C. IR (thin film) 3065, 2942, 2782, 1700, 1664, 1609, 1590, 1563, 1539, 1499 cm−1; 1H NMR (300 MHz, DMSO-d6) δ 8.46 (d, J=8.6 Hz, 1H), 8.28 (s, 1H), 8.13 (s, 1H), 7.89 (d, J=8.5 Hz, 1H), 7.58 (s, 1H), 4.85 (m, 2H), 3.95 (s, 3H), 3.06 (s, 1H), 2.88 (m, 2H), 2.03 (m, 2H), 1.10 (d, J=5.6 Hz, 6H); MALDIMS m/z (rel intensity) 412 (MH+, 100); HRESIMS calcd for C22H22ClN3O3(MH+) 412.1428, found 412.1420; HPLC purity, 100.00% (MeOH—H2O, 85:15).
3-Chloro-9-methoxy-6-(3-(methylamino)propyl)-5H-pyrido[3′,2′:4,5]cyclopenta[1,2-c]isoquinoline-5,11(6H)-dione (16g)
The reaction to prepare this compound was performed in a 45 mL pressure vessel and the product was isolated as red powder (0.038 g, 77%): mp 274-275° C. IR (thin film) 2941, 2777, 1699, 1669, 1608, 1565, 1535, 1500 cm−1; 1H NMR (300 MHz, DMSO-d6) δ 8.48 (d, J=8.7 Hz, 1H), 8.29 (d, J=2.7 Hz, 1H), 8.15 (d, J=2.4 Hz, 1H), 7.90 (d, J=8.6 Hz, 1H), 7.60 (d, J=2.7 Hz, 1H), 4.85 (m, 2H), 3.95 (s, 3H), 3.33 (s, 3H), 2.91 (m, 2H), 2.05 (m, 2H); MALDIMS m/z (rel intensity) 384 (MH+, 100); HRESIMS calcd for C20H18ClN3O3(MH+) 384.1115, found 384.1108; HPLC purity, 95.81% (MeOH—H2O, 85:15).
3-Chloro-9-methoxy-6-(3-(ethylamino)propyl)-5H-pyrido[3′,2′:4,5]cyclopenta[1,2-c]isoquinoline-5,11(6H)-dione (16h)
The reaction to prepare this compound was performed in a 45 mL pressure vessel and the product was isolated as red powder (0.04 g, 80%): mp 264-266° C. IR (thin film) 2945, 2756, 2497, 1710, 1661, 1608, 1566, 1537, 1499 cm−1; 1H NMR (300 MHz, DMSO-d6) δ 8.47 (d, J=8.7 Hz, 1H), 8.28 (d, J=2.7 Hz, 1H), 8.14 (d, J=2.3 Hz, 1H), 7.88 (dd, J=8.7, 2.3 Hz, 1H), 7.57 (d, J=2.7 Hz, 1H), 4.84 (d, J=7.0 Hz, 2H), 3.96 (s, 3H), 2.83 (t, J=7.4 Hz, 2H), 2.73 (q, J=7.3 Hz, 2H), 2.00 (m, 2H), 1.06 (t, J=7.2 Hz, 3H); MALDIMS m/z (rel intensity) 398 (MH+, 100); HRESIMS calcd for C21H20ClN3O3(MH+) 398.1272, found 398.1258; HPLC purity, 97.07% (MeOH—H2O, 85:15).
3-Chloro-9-methoxy-6-(3-(pyrrolidin-1-yl)propyl)-5H-pyrido[3′,2′:4,5]cyclopenta[1,2-c]isoquinoline-5,11(6H)-dione (16i)
This compound was isolated as red powder (0.045 g, 83%): mp 242-244° C. IR (thin film) 3063, 2934, 2787, 1699, 1664, 1608, 1565, 1537, 1501 cm−1; 1H NMR (300 MHz, DMSO-d6) δ 8.45 (d, J=8.7 Hz, 1H), 8.27 (d, J=2.7 Hz, 1H), 8.12 (d, J=2.2 Hz, 1H), 7.86 (dd, J=8.7, 2.3 Hz, 1H), 7.55 (d, J=2.8 Hz, 1H), 4.90 (m, 2H), 4.11 (m, 2H), 3.95 (s, 3H), 3.16 (d, J=5.1 Hz, 4H), 1.96 (m, 2H), 1.57 (m, 4H); MALDIMS m/z (rel intensity) 424 (MH+, 100); HRESIMS calcd for C23H22ClN3O3(MH+) 424.1428, found 424.1419; HPLC purity, 96.93% (MeOH—H2O, 85:15).
3-Chloro-9-methoxy-6-(3-(4-methylpiperazin-1-yl)propyl)-5H-pyrido[3′,2′:4,5]cyclopenta[1,2-c]isoquinoline-5,11(6H)-dione (16j)
This compound was isolated as red powder (0.042 g, 72%): mp 253-255° C. IR (thin film) 2931, 2789, 1699, 1666, 1607, 1565, 1537, 1501 cm1; 1H NMR (300 MHz, DMSO-d6) δ 8.42 (d, J=8.8 Hz, 1H), 8.23 (d, J=2.8 Hz, 1H), 8.08 (d, J=2.2 Hz, 1H), 7.83 (dd, J=8.7, 2.3 Hz, 1H), 7.51 (d, J=2.7 Hz, 1H), 4.89 (m, 2H), 3.93 (s, 3H), 2.37 (m, 2H), 2.12 (m, 8H), 1.96 (s, 3H), 1.87 (m, 2H); MALDIMS m/z (rel intensity) 453 (MH+, 100); HRESIMS calcd for C24H25ClN4O3(MH+) 453.1694, found 453.1686; HPLC purity, 95.88% (MeOH—H2O, 85:15).
General Procedures for Preparation of Compounds 17a-i.
Compound 15b (50 mg, 0.134 mmol), K2CO3 (0.05 g, 1 mmol), NaI (40 mg) and the appropriate amine (10 eq) were mixed in DMF (5 mL). The mixture was stirred overnight at 90° C. and then cooled to room temperature. Water (30 mL) was added to the reaction flask and then the mixture was extracted with EtOAc (3×10 mL). The combined organic layers were washed with water (3×15 mL) and brine (15 mL), dried over Na2SO4, and evaporated to give a red residue. The residue was purified by silica gel column chromatography (MeOH—CHCl3, 5:95) to yield compounds 17a-i. Compounds 17f-h were prepared using the same procedures but the reaction was performed in a 15 mL pressure vessel.
3-Fluoro-9-methoxy-6-(3-morpholinopropyl)-5H-pyrido[3′,2′:4,5]cyclopenta[1,2c]-isoquinoline-5,11(6H)-dione (17a)
This compound was isolated as a dark red solid (46 mg, 84%): mp 175-178° C. IR (thin film) 3070, 2856, 2806, 1698, 1658, 1593, 1548, 1511 cm1; 1H NMR (300 MHz, DMSO-d6) δ 8.50 (dd, J=9.0, 5.4 Hz, 1H), 8.22 (d, J=2.8 Hz, 1H), 7.86 (dd, J=9.5, 2.7 Hz, 1H), 7.72 (td, J=8.8, 2.8 Hz, 1H), 7.52 (d, J=2.7 Hz, 1H), 4.88 (t, J=7.4 Hz, 2H), 3.94 (s, 3H), 3.34 (m, 4H), 2.40 (t, J=6.4 Hz, 2H), 2.20 (m, 4H), 1.91 (d, J=6.7 Hz, 2H); MALDI m/z (rel intensity) 424 (MH+, 100); HRMS-ESI m/z MH+ calcd for C23H23FN3O4, 424.1673; found, 424.1665; HPLC purity: 96.06% (MeOH—H2O, 85:15).
6-(3-(1H-Imidazol-1-yl)propyl)-3-fluoro-9-methoxy-5H-pyrido[3′,2′:4,5]-cyclo-penta[1,2-c]isoquinoline-5,11(6H)-dione (17b)
This compound was isolated as a dark red solid (37.5 mg, 69%): mp 176-180° C. IR (thin film) 3543, 2969, 1707, 1657, 1592, 1572, 1508 cm−1; 1H NMR (300 MHz, DMSO-d6) δ 8.53 (dd, J=9.0, 5.4 Hz, 1H), 8.15 (d, J=2.8 Hz, 1H), 7.95-7.69 (m, 2H), 7.55 (d, J=2.8 Hz, 1H), 7.28 (s, 1H), 6.98 (s, 1H), 4.81 (t, J=7.4 Hz, 2H), 4.14 (t, J=7.0 Hz, 2H), 3.95 (s, 3H), 2.27-2.16 (m, 2H); MALDI m/z (rel intensity) 405 (MH+, 100); HRMS-ESI m/z MH+ calcd for C22H18FN4O3, 405.1363; found, 405.1357; HPLC purity: 96.60% (MeOH—H2O, 85:15).
6-(3-((4,5-Dihydrothiazol-2-yl)amino)propyl)-3-fluoro-9-methoxy-5H-pyrido[3′,2′:4,5]-cyclopenta-[1,2-c]isoquinoline-5,11(6H)-dione (17c)
This compound was isolated as a dark red solid (25 mg, 43%): mp 226-250° C. IR (thin film) 2937, 1699, 1657, 1593, 1549, 1510 cm−1; 1H NMR (300 MHz, DMSO-d6) δ 8.56 (d, J=5.5 Hz, 1H), 8.25 (d, J=2.7 Hz, 1H), 7.90 (d, J=9.5 Hz, 1H), 7.79 (d, J=8.8 Hz, 1H), 7.59 (d, J=2.7 Hz, 1H), 4.85 (m, 2H), 4.03 (d, J=7.3 Hz, 2H), 3.96 (s, 3H), 3.68 (m, 2H), 3.51 (d, J=7.4 Hz, 2H), 2.09 (m, 2H); ESIMS m/z (rel intensity) 439 (MH+, 100); HRMS-ESI m/z MH+ calcd for C22H20FN4O3S, 439.1240; found, 439.1233; HPLC purity: 95.01% (MeOH—H2O, 85:15).
3-Fluoro-6-(3-(4-hydroxypiperidin-1-yl)propyl)-9-methoxy-5H-pyrido[3′,2′:4,5]-cyclopenta[1,2-c]-isoquinoline-5,11(6H)-dione (17d)
This compound was isolated as a dark red solid (30 mg, 50%): mp 205-209° C. IR (thin film) 3263, 2942, 1668, 1591, 1551, 1510 cm−1; 1H NMR (300 MHz, DMSO-d6) δ 8.54-8.44 (m, 1H), 8.24 (d, J=2.1 Hz, 1H), 7.87 (d, J=6.9 Hz, 1H), 7.73 (s, 1H), 7.53 (s, 1H), 4.88 (m, 2H), 4.47 (m, 1H), 3.94 (s, 3H), 2.57 (m, 1H), 2.40 (m, 2H), 1.89 (m, 4H), 1.50 (m, 2H), 1.07 (m, 2H); ESIMS m/z (rel intensity) 437 (MH+, 100); HRMS-ESI m/z MH+ calcd for C24H24FN3O4, 437.1989; found, 437.1981; HPLC purity: 95.24% (MeOH—H2O, 85:15).
3-Fluoro-9-methoxy-6-(3-(piperazin-1-yl)propyl)-5H-pyrido[3′,2′:4,5]cyclopenta-[1,2-c]isoquinoline-5,11(6H)-dione (17e)
This compound was isolated as a dark red solid (44 mg, 78%): mp 237-239° C. IR (thin film) 2936, 2808, 1699, 1658, 1593, 1550, 1510 cm−1; 1H NMR (300 MHz, DMSO-d6) δ 8.51 (dd, J=9.0, 5.4 Hz, 1H), 8.23 (d, J=2.8 Hz, 1H), 7.86 (dd, J=9.5, 2.8 Hz, 1H), 7.73 (td, J=8.8, 2.8 Hz, 1H), 7.53 (d, J=2.8 Hz, 1H), 4.87 (t, J=7.3 Hz, 2H), 3.94 (s, 3H), 2.69 (m, 4H), 2.43 (t, J=6.5 Hz, 2H), 2.30 (m, 4H), 1.90 (t, J=6.9 Hz, 2H); MALDI m/z (rel intensity) 423 (MH+, 100); HRMS-ESI m/z MH+ calcd for C23H23N4O3F, 423.1833; found, 423.1824; HPLC purity: 95.28% (MeOH—H2O, 85:15).
3-Fluoro-6-(3-(isopropylamino)propyl)-9-methoxy-5H-pyrido[3′,2′:4,5]cyclopenta[1,2-c]isoquinoline-5,11(6H)-dione (17f)
This compound was isolated as a dark red solid (41 mg, 75%): mp 285-292° C. IR (thin film) 2714, 1705, 1659, 1588, 1552, 1511 cm−1; 1H NMR (300 MHz, DMSO-d6) δ 8.52 (dd, J=8.9, 5.5 Hz, 1H), 8.36-8.22 (m, 2H), 7.89 (dd, J=9.4, 2.8 Hz, 1H), 7.75 (td, J=8.7, 2.7 Hz, 1H), 7.57 (t, J=2.5 Hz, 1H), 4.85 (t, J=7.1 Hz, 2H), 3.95 (s, 3H), 3.05 (m, 1H), 2.86 (d, J=7.8 Hz, 2H), 2.04 (m, 2H), 1.10 (d, J=6.5 Hz, 6H); MALDI m/z (rel intensity) 396 (MH+, 100); HRMS-ESI m/z MH+ calcd for C22H23FN3O3, 396.1724; found, 396.1719; HPLC purity: 96.70% (MeOH—H2O, 85:15).
3-Fluoro-9-methoxy-6-(3-(methylamino)propyl)-5H-pyrido[3′,2′:4,5]cyclopenta[1,2-c]isoquinoline-5,11(6H)-dione (17g)
This compound was isolated as a dark red solid (31 mg, 63%): mp 259-265° C. IR (thin film) 1667, 1554, 1513, 1484, 1454 cm−1; 1H NMR (300 MHz, DMSO-d6) δ 8.56 (dd, J=8.9, 5.3 Hz, 1H), 8.29 (d, J=2.7 Hz, 1H), 7.92 (dd, J=9.4, 2.7 Hz, 1H), 7.86-7.75 (m, 1H), 7.61 (d, J=2.7 Hz, 1H), 4.87 (m, 2H), 3.96 (s, 3H), 3.00 (t, J=7.7 Hz, 2H), 2.54 (s, 3H), 2.11 (m, 2H); ESIMS m/z (rel intensity) 368 (MH+, 100); HRMS-ESI m/z MH+ calcd for C20H19FN3O3, 368.1411; found, 368.1402; HPLC purity: 95.05% (MeOH—H2O, 85:15).
6-(3-(Ethylamino)propyl)-3-fluoro-9-methoxy-5H-pyrido[3′,2′:4,5]cyclopenta[1,2-c]isoquinoline-5,11(6H)-dione (17h)
This compound was isolated as a dark red solid (18.8 mg, 37.1%): mp 286-288° C. IR (thin film) 3451, 2924, 2851. 2783, 1700, 1671, 1616, 1592, 1573, 1552, 1511 cm−1; 1H NMR (300 MHz, DMSO-d6) δ 8.55 (dd, J=9.0, 5.5 Hz, 1H), 8.27 (d, J=2.8 Hz, 1H), 7.91 (dd, J=9.4, 2.8 Hz, 1H), 7.84-7.72 (m, 1H), 7.60 (d, J=2.8 Hz, 1H), 4.86 (t, J=6.9 Hz, 2H), 3.96 (s, 3H), 3.04-2.82 (m, 4H), 2.10 (d, J=8.2 Hz, 2H), 1.26-1.08 (m, 3H); MALDI m/z (rel intensity) 382 (MH+, 100); HRMS-ESI m/z MH+ calcd for C21H21FN3O3, 382.1567; found, 382.1558; HPLC purity: 95.58% (MeOH—H2O, 85:15).
3-Fluoro-9-methoxy-6-(3-(pyrrolidin-1-yl)propyl)-5H-pyrido[3′,2′:4,5]cyclopenta[1,2-c]isoquinoline-5,11(6H)-dione (17i)
This compound was isolated as a dark red solid (20 mg, 37%): mp 157-162° C. IR (thin film) 1702, 1663, 1614, 1572, 1550, 1510, 1480, 1446, 1434 cm−1; 1H NMR (300 MHz, DMSO-d6) δ 8.51 (dd, J=8.9, 5.4 Hz, 1H), 8.26 (t, J=2.6 Hz, 1H), 7.88 (dd, J=9.5, 2.8 Hz, 1H), 7.74 (td, J=8.7, 2.8 Hz, 1H), 7.54 (t, J=2.5 Hz, 1H), 4.90 (t, J=6.9 Hz, 2H), 3.95 (s, 3H), 2.73 (m, 4H), 1.98 (m, 3H), 1.60 (m, 5H); ESIMS m/z (rel intensity) 408 (MH+, 100); HRMS-ESI m/z MH+ calcd for C27H23FN3O3, 408.1724; found, 408.1717; HPLC purity: 95.08%. (MeOH—H2O, 85:15).
Topoisomerase I-Mediated DNA Cleavage Reactions
(Dexheimer, et al., Nat. Protoc. 2008, 3, 1736-1750). A 3′-[32P]-labeled 117-bp DNA oligonucleotide was prepared as previously described. The oligonucleotide contains previously identified Top1 cleavage sites in 161-bp pBluescript SK(−) phagemid DNA. Approximately 2 nM radiolabeled DNA substrate was incubated with recombinant Top1 in 20 μL of reaction buffer [10 mM Tris-HCl (pH 7.5), 50 mM KCl, 5 mM MgCl2, 0.1 mM EDTA, and 15 μg/mL BSA] at 25° C. for 20 min in the presence of various concentrations of test compounds. The reactions were terminated by adding SDS (0.5% final concentration) followed by the addition of two volumes of loading dye (80% formamide, 10 mM sodium hydroxide, 1 mM sodium EDTA, 0.1% xylene cyanol, and 0.1% bromophenol blue). Aliquots of each reaction mixture were subjected to 20% denaturing PAGE. Gels were dried and visualized by using a phosphoimager and ImageQuant software (Molecular Dynamics). Cleavage sites are numbered to reflect actual sites on the 117 bp oligonucleotide (Antony, et al., Nucleic Acids Res. 2007, 35, 4474-4484).
Recombinant Tdp1 Assay
(Nguyen, et al., J. Med. Chem. 2012, 55, 4457-4478). A 5′-[32P]-labeled single-stranded DNA oligonucleotide containing a 3′-phosphotyrosine (N14Y) was incubated at 1 nM with 10 pM recombinant TDP1 in the absence or presence of inhibitor for 15 min at room temperature in the LMP1 assay buffer containing 50 mM Tris HCl, pH 7.5, 80 mM KCl, 2 mM EDTA, 1 mM DTT, 40 μg/mL BSA, and 0.01% Tween-20. Reactions were terminated by the addition of 1 volume of gel loading buffer [99.5% (v/v) formamide, 5 mM EDTA, 0.01% (w/v) xylene cyanol, and 0.01% (w/v) bromophenol blue]. Samples were subjected to a 16% denaturing PAGE with multiple loadings at 12-min intervals. Gels were dried and exposed to a PhosphorImager screen (GE Healthcare). Gel images were scanned using a Typhoon 8600 (GE Healthcare), and densitometry analyses were performed using the ImageQuant software (GE Healthcare).
Recombinant Tdp2 Assay
(Gao, et al., J. Biol. Chem. 2012, 287, 30842-30852). TDP2 reactions were carried out as described previously with the following modifications. The 18-mer single-stranded oligonucleotide DNA substrate (TY18, α32P-cordycepin-3′-labeled) was incubated at 1 nM with 25 pM recombinant human TDP2 in the absence or presence of inhibitor for 15 min at room temperature in the LMP2 assay buffer containing 50 mM Tris-HCl, pH 7.5, 80 mM KCl, 5 mM MgCl2, 0.1 mM EDTA, 1 mM DTT, 40 μg/mL BSA, and 0.01% Tween 20. Reactions were terminated and treated similarly to whole-cell extract and recombinant Tdp1 reactions (see previous paragraph).
Those skilled in the art will recognize that numerous modifications can be made to the specific implementations described above. The implementations should not be limited to the particular limitations described. Other implementations may be possible.
While the inventions have been illustrated and described in detail in the drawings and foregoing description, the same is to be considered as illustrative and not restrictive in character, it being understood that only certain embodiments have been shown and described and that all changes and modifications that come within the spirit of the invention are desired to be protected.

Claims (9)

What is claimed is:
1. A compound of formula (II)
Figure US10875860-20201229-C00012
or a pharmaceutically acceptable salt thereof, wherein m is 3, R1 is 3-Cl or 3-F, and R2 is selected from the group consisting of
Figure US10875860-20201229-C00013
2. The compound or salt of claim 1 wherein R1 is 3-F.
3. The compound or salt of claim 1 wherein R1 is 3-Cl.
4. The compound or salt of claim 2 wherein R2 is MeHN—, EtHN—, or i-PrHN—.
5. The compound or salt of claim 3 wherein R2 is MeHN—, EtHN—, or i-PrHN—.
6. A pharmaceutical composition comprising the compound of claim 1, or a pharmaceutically acceptable salt thereof, and one or more pharmaceutically acceptable carriers, diluents, and excipients.
7. A method for treating a patient with a cancer selected from the group consisting of lung cancer, colon cancer, a CNS cancer, melanoma, ovarian cancer, renal cancer, prostate cancer, and breast cancer, the method comprising the step of administering a therapeutically effective amount of the pharmaceutical composition of claim 6 to the patient in need of relief from said cancer.
8. A method for treating a patient with a cancer selected from the group consisting of lung cancer, colon cancer, a CNS cancer, melanoma, ovarian cancer, renal cancer, prostate cancer, and breast cancer, the method comprising the step of administering a therapeutically effective amount of the compound or salt of claim 1, together with one or more pharmaceutically acceptable carriers, diluents, and excipients, to the patient in need of relief from said cancer.
9. A method for treating a patient with a cancer selected from the group consisting of lung cancer, colon cancer, a CNS cancer, melanoma, ovarian cancer, renal cancer, prostate cancer, and breast cancer, the method comprising the step of administering a therapeutically effective amount of the compound or salt of claim 1, and a therapeutically effective amount of one or more other compounds of the same or different mode of action, together with one or more pharmaceutically acceptable carriers, diluents, and excipients, to the patient in need of relief from said cancer.
US16/471,945 2016-12-22 2017-12-19 Azaindenoisoquinoline compounds and uses thereof Active US10875860B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/471,945 US10875860B2 (en) 2016-12-22 2017-12-19 Azaindenoisoquinoline compounds and uses thereof

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201662437777P 2016-12-22 2016-12-22
PCT/US2017/067206 WO2018118852A1 (en) 2016-12-22 2017-12-19 Azaindenoisoquinoline compounds and uses thereof
US16/471,945 US10875860B2 (en) 2016-12-22 2017-12-19 Azaindenoisoquinoline compounds and uses thereof

Publications (2)

Publication Number Publication Date
US20200095243A1 US20200095243A1 (en) 2020-03-26
US10875860B2 true US10875860B2 (en) 2020-12-29

Family

ID=62627773

Family Applications (1)

Application Number Title Priority Date Filing Date
US16/471,945 Active US10875860B2 (en) 2016-12-22 2017-12-19 Azaindenoisoquinoline compounds and uses thereof

Country Status (6)

Country Link
US (1) US10875860B2 (en)
EP (1) EP3558993A4 (en)
JP (1) JP7182548B2 (en)
CN (1) CN110167936A (en)
CA (1) CA3047992A1 (en)
WO (1) WO2018118852A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020023700A2 (en) * 2018-07-25 2020-01-30 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services OXYNITIDINE DERIVATIVES USEFUL AS INHIBITORS OF TOPOISOMERASE IB (Top1) AND TYROSYL-DNA PHOSPHODIESTERASE 1 (Tdp1)

Citations (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5023261A (en) 1988-08-30 1991-06-11 Kissei Pharmaceutical Co., Ltd. Acylated benzofuro[3,2-c]quinoline compounds with utility as treatments for osteoporosis
WO2000021537A1 (en) 1998-10-14 2000-04-20 Purdue Research Foundation Novel indenoisoquinolines as antineoplastic agents
WO2004100891A2 (en) 2003-05-12 2004-11-25 Purdue Research Foundation Cytotoxic indeno and isoindoloisoquinolones
US6828319B2 (en) 2001-08-31 2004-12-07 Inotek Pharmaceuticals Corporation Substituted indeno[1,2-c]isoquinoline derivatives and methods of use thereof
US20050010046A1 (en) 2001-11-14 2005-01-13 Lavoie Edmond J. Topoisomerase poison agents
WO2005089294A2 (en) 2004-03-17 2005-09-29 Purdue Research Foundation Synthesis of indenoisoquinoliniums and methods of use
US20060025595A1 (en) 2004-05-07 2006-02-02 Cushman Mark S Synthesis of indenoisoquinolines
WO2007059008A2 (en) 2005-11-14 2007-05-24 Purdue Research Foundation N-substituted indenoisoquinolines and syntheses thereof
US20080090831A1 (en) 2001-11-14 2008-04-17 Rutgers, The State University Of New Jersey Cytotoxic agents
WO2008076767A1 (en) 2006-12-13 2008-06-26 Purdue Research Foundation Chemotherapeutic flavonoids, and syntheses thereof
WO2009140467A1 (en) 2008-05-15 2009-11-19 Purdue Research Foundation Oxobenzindolizinoquinolines and uses thereof
WO2011094416A1 (en) 2010-01-27 2011-08-04 Purdue Research Foundation Substituted norindenoisoquinolines, syntheses thereof, and methods of use
WO2012024437A1 (en) 2010-08-17 2012-02-23 Purdue Research Foundation Substituted dibenzonaphthyridines, pharmaceutical uses thereof and processes therfor
WO2012162513A2 (en) 2011-05-25 2012-11-29 Purdue Research Foundation Alcohol-, diol-, and carbohydrate-substituted indenoisoquinolines as topoisomerase i inhibitors
US20130345252A1 (en) 2012-04-13 2013-12-26 Purdue Research Foundation Synthesis and use of dual tyrosyl-dna phosphodiesterase i (tdp1)- topoisomerase i (top1) inhibitors
US20140018360A1 (en) 2012-07-13 2014-01-16 Mark S. Cushman Azaindenoisoquinoline topoisomerase i inhibitors
US20140187547A1 (en) 2012-07-13 2014-07-03 Purdue Research Foundation Azaindenoisoquinoline topoisomerase i inhibitors
WO2015069766A1 (en) 2013-11-06 2015-05-14 Purdue Research Foundation Dupa-indenoisoquinoline conjugates
US20150210686A1 (en) 2012-08-08 2015-07-30 Merck Patent Gmbh (aza-)isoquinolinone derivatives
US20150299246A1 (en) 2005-11-14 2015-10-22 Purdue Research Foundation N-substituted indenoisoquinolines and syntheses thereof
US20160318946A1 (en) 2011-05-25 2016-11-03 Purdue Research Foundation Alcohol-, diol-, and carbohydrate-substituted indenoisoquinolines as topoisomerase i inhibitors

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2010520220A (en) * 2007-02-28 2010-06-10 イノテック ファーマシューティカルズ コーポレイション Indenoisoquinolinone analogs and methods of use thereof

Patent Citations (51)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5023261A (en) 1988-08-30 1991-06-11 Kissei Pharmaceutical Co., Ltd. Acylated benzofuro[3,2-c]quinoline compounds with utility as treatments for osteoporosis
WO2000021537A1 (en) 1998-10-14 2000-04-20 Purdue Research Foundation Novel indenoisoquinolines as antineoplastic agents
US6509344B1 (en) 1998-10-14 2003-01-21 The United States Of America As Represented By The Department Of Health And Human Services Indenoisoquinolines as antineoplastic agents
US6828319B2 (en) 2001-08-31 2004-12-07 Inotek Pharmaceuticals Corporation Substituted indeno[1,2-c]isoquinoline derivatives and methods of use thereof
US20080090831A1 (en) 2001-11-14 2008-04-17 Rutgers, The State University Of New Jersey Cytotoxic agents
US20050010046A1 (en) 2001-11-14 2005-01-13 Lavoie Edmond J. Topoisomerase poison agents
WO2004100891A2 (en) 2003-05-12 2004-11-25 Purdue Research Foundation Cytotoxic indeno and isoindoloisoquinolones
US20060247211A1 (en) 2003-05-12 2006-11-02 Cushman, Mark S., Pommier, Yves G. Cytotoxic indeno and isoindoloisoquinolones
US7312228B2 (en) 2003-05-12 2007-12-25 Purdue Research Foundation Cytotoxic indeno and isoindoloisoquinolones
WO2005089294A2 (en) 2004-03-17 2005-09-29 Purdue Research Foundation Synthesis of indenoisoquinoliniums and methods of use
US7781445B2 (en) 2004-03-17 2010-08-24 Purdue Research Foundation Synthesis of indenoisoquinoliniums and methods of use
US20080242692A1 (en) 2004-03-17 2008-10-02 Cushman Mark S Synthesis of Indenoisoquinoliniums and Methods of Use
US20060025595A1 (en) 2004-05-07 2006-02-02 Cushman Mark S Synthesis of indenoisoquinolines
US7495100B2 (en) 2004-05-07 2009-02-24 Purdue Research Foundation Synthesis of indenoisoquinolines
US9399660B2 (en) 2005-11-14 2016-07-26 Purdue Research Foundation N-substituted indenoisoquinolines and syntheses thereof
US20120101119A1 (en) 2005-11-14 2012-04-26 Cushman Mark S N-substituted indenoisoquinolines and syntheses thereof
US8829022B2 (en) 2005-11-14 2014-09-09 Purdue Research Foundation N-substituted indenoisoquinolines and syntheses thereof
US20150299246A1 (en) 2005-11-14 2015-10-22 Purdue Research Foundation N-substituted indenoisoquinolines and syntheses thereof
US20150218207A1 (en) 2005-11-14 2015-08-06 Purdue Research Foundation N-substituted indenoisoquinolines and syntheses thereof
US8053443B2 (en) 2005-11-14 2011-11-08 Purdue Research Foundation N-substituted indenoisoquinolines and syntheses thereof
US9796753B2 (en) 2005-11-14 2017-10-24 Purdue Research Foundation N-substituted indenoisoquinolines and syntheses thereof
US20140336188A1 (en) 2005-11-14 2014-11-13 Purdue Research Foundation N-substituted indenoisoquinolines and syntheses thereof
US20160229888A1 (en) 2005-11-14 2016-08-11 Purdue Research Foundation N-substituted indenoisoquinolines and syntheses thereof
US20080318995A1 (en) 2005-11-14 2008-12-25 Cushman Mark S N-Substituted Indenoisoquinolines and Syntheses Thereof
WO2007059008A2 (en) 2005-11-14 2007-05-24 Purdue Research Foundation N-substituted indenoisoquinolines and syntheses thereof
US9388211B2 (en) 2005-11-14 2016-07-12 Purdue Research Foundation N-substituted indenoisoquinolines and syntheses thereof
US9217010B2 (en) 2005-11-14 2015-12-22 Purdue Research Foundation N-substituted indenoisoquinolines and syntheses thereof
WO2008076767A1 (en) 2006-12-13 2008-06-26 Purdue Research Foundation Chemotherapeutic flavonoids, and syntheses thereof
WO2009140467A1 (en) 2008-05-15 2009-11-19 Purdue Research Foundation Oxobenzindolizinoquinolines and uses thereof
US20120302563A1 (en) 2010-01-27 2012-11-29 Purdue Research Foundation Substituted norindenoisoquinolines, syntheses thereof, and methods of use
US9206193B2 (en) 2010-01-27 2015-12-08 Purdue Research Foundation Substituted norindenoisoquinolines, syntheses thereof, and methods of use
WO2011094416A1 (en) 2010-01-27 2011-08-04 Purdue Research Foundation Substituted norindenoisoquinolines, syntheses thereof, and methods of use
US9073920B2 (en) 2010-08-17 2015-07-07 Purdue Research Foundation Substituted dibenzonaphthyridines, pharmaceutical uses thereof and processes therfor
WO2012024437A1 (en) 2010-08-17 2012-02-23 Purdue Research Foundation Substituted dibenzonaphthyridines, pharmaceutical uses thereof and processes therfor
US9328073B2 (en) 2011-05-25 2016-05-03 Purdue Research Foundation Alcohol-, diol-, and carbohydrate-substituted indenoisoquinolines as topoisomerase I inhibitors
US20150148370A1 (en) 2011-05-25 2015-05-28 Purdue Research Foundation Alcohol-, diol-, and carbohydrate-substituted indenoisoquinolines as topoisomerase i inhibitors
US9682990B2 (en) 2011-05-25 2017-06-20 Purdue Research Foundation Alcohol-, diol-, and carbohydrate-substituted indenoisoquinolines as topoisomerase I inhibitors
US20160318946A1 (en) 2011-05-25 2016-11-03 Purdue Research Foundation Alcohol-, diol-, and carbohydrate-substituted indenoisoquinolines as topoisomerase i inhibitors
WO2012162513A2 (en) 2011-05-25 2012-11-29 Purdue Research Foundation Alcohol-, diol-, and carbohydrate-substituted indenoisoquinolines as topoisomerase i inhibitors
US20130345252A1 (en) 2012-04-13 2013-12-26 Purdue Research Foundation Synthesis and use of dual tyrosyl-dna phosphodiesterase i (tdp1)- topoisomerase i (top1) inhibitors
US20160081999A1 (en) 2012-04-13 2016-03-24 Purdue Research Foundation SYNTHESIS AND USE OF DUAL TYROSYL-DNA PHOSPHODIESTERASE I (Tdp1) - TOPOISOMERASE I (Top1) INHIBITORS
US9175002B2 (en) 2012-04-13 2015-11-03 Purdue Research Foundation Synthesis and use of dual tyrosyl-DNA phosphodiesterase I (Tdp1)—topoisomerase I (Top1) inhibitors
US9402842B2 (en) 2012-04-13 2016-08-02 Purdue Research Foundation Synthesis and use of dual tyrosyl-DNA phosphodiesterase I (Tdp1)—topoisomerase I (Top1) inhibitors
US8912213B2 (en) 2012-04-13 2014-12-16 Purdue Research Foundation Synthesis and use of dual tyrosyl-DNA phosphodiesterase I (TDP1)- topoisomerase I (TOP1) inhibitors
US20150133445A1 (en) 2012-04-13 2015-05-14 Purdue Research Foundation SYNTHESIS AND USE OF DUAL TYROSYL-DNA PHOSPHODIESTERASE I (Tdp1) - TOPOISOMERASE I (Top1) INHIBITORS
US20140187547A1 (en) 2012-07-13 2014-07-03 Purdue Research Foundation Azaindenoisoquinoline topoisomerase i inhibitors
US8686146B2 (en) 2012-07-13 2014-04-01 Purdue Research Foundation Azaindenoisoquinoline topoisomerase I inhibitors
US20140018360A1 (en) 2012-07-13 2014-01-16 Mark S. Cushman Azaindenoisoquinoline topoisomerase i inhibitors
US9034870B2 (en) 2012-07-13 2015-05-19 Purdue Research Foundation Azaindenoisoquinoline topoisomerase I inhibitors
US20150210686A1 (en) 2012-08-08 2015-07-30 Merck Patent Gmbh (aza-)isoquinolinone derivatives
WO2015069766A1 (en) 2013-11-06 2015-05-14 Purdue Research Foundation Dupa-indenoisoquinoline conjugates

Non-Patent Citations (29)

* Cited by examiner, † Cited by third party
Title
Antony et al., Molecular Pharmacology, 67: 523-530. *
Arpicco et al., Anticancer Prodrugs: An Overview of Major Strategies and Recent Developments, Current topics in medical chemistry, Jun. 2011, pp. 2345-2381.
Beck Daniel E et al: "Synthesis and biological evaluation of new fluorinated and chlorinated indenoisoquinoline topoisomerase I poisons", Bioorganic & Medicinal Chemistry, vol. 24, No. 7, Feb. 9, 2016, pp. 1469-1479, XP029459043.
BECK DANIEL E.; LV WEI; ABDELMALAK MONICA; PLESCIA CAROLINE B.; AGAMA KELI; MARCHAND CHRISTOPHE; POMMIER YVES; CUSHMAN MARK: "Synthesis and biological evaluation of new fluorinated and chlorinated indenoisoquinoline topoisomerase I poisons", BIOORGANIC & MEDICINAL CHEMISTRY, ELSEVIER, NL, vol. 24, no. 7, 9 February 2016 (2016-02-09), NL, pages 1469 - 1479, XP029459043, ISSN: 0968-0896, DOI: 10.1016/j.bmc.2016.02.015
Beck et al. ‘Discovery of Potent Indenoisoquinoline Topoisomerase I Poisons Lacking the Nitro Toxicophore’, Journal of medicinal chemistry, 2015, vol. 58.9, pp. 3997-4015. p. 3997, col. 1, para 2; p. 3998, col. 1, para 1; p. 4001, Scheme 8; p. 4003, Table 2.
Beck, Daniel E., et al. "Investigation of the Structure-Activity Relationships of Aza—A-Ring Indenoisoquinoline Topoisomerase I Poisons." Journal of medicinal chemistry 59.8 (2016): 3840-3853.
Cancer and Metastasis Reviews (1998), 17(1), 91-106. *
Cinelli et al., Identification, Sythesis, and Biological Evaluation of Metabolites of the Experimental Cancer Treatment Drugs Indotecan (LMP400) and Indimitecan (LMP776) and Investigation of Isomerically Hydroxylated Indenoisoquinoline Analogues as Topoisomerase I Poisons, Journal of Medicinal Chemistry, 2012, pp. 10844-10862, American Chemical Society.
Elsayed, Mohamed SA, et al. "Design and synthesis of chlorinated and fluorinated 7-azaindenoisoquinolines as potent cytotoxic anticancer agents that inhibit topoisomerase I." Journal of medicinal chemistry 60.13 (2017): 5364-5376.
International Search Report, PCT/US17/16331, dated Apr. 28, 2017.
International Search Report, PCT/US2017/022389, dated Jun. 9, 2017.
International Search Report, PCT/US2017/067206, dated Apr. 24, 2018.
Jung "Prednisolone 21-sulfate sodium: a colon-specific pro-drug of prednisolone" 2003, 55, 1075-1082.
Kiselev et al. "Optimization of the Lactam Side Chain of 7-Azaindenoisoquinoline Topoisomerase I Inhibitors and Mechanism of Action Studies in Cancer Cells" Journal of Medicinal Chemistry. Feb. 6, 2014 (Feb. 6, 2014) vol. 57, p. 1289-1298; p. 1290.
Kiselev, Evgeny, et al. "7-azaindenoisoquinolines as topoisomerase I inhibitors and potential anticancer agents." Journal of medicinal chemistry 54.17 (2011): 6106-6116.
Kiselev, Evgeny, et al. "Azaindenoisoquinolines as topoisomerase I inhibitors and potential anticancer agents: a systematic study of structure-activity relationships." Journal of medicinal chemistry 55.4 (2012): 1682-1697.
Kiselev, Evgeny, et al. "Optimization of the lactam side chain of 7-azaindenoisoquinoline topoisomerase I inhibitors and mechanism of action studies in cancer cells." Journal of medicinal chemistry 57.4 (2014): 1289-1298.
Paull, K. D. et al., Display and Analysis of Patterns of Differential Activity of Drugs Against Human Tumor Cell Lines: Development of Mean Graph and COMPARE Algorithm, J. Natl. Cancer Inst., Jul. 19, 1989, pp. 1088-1091, vol. 81, No. 14.
Pommier, Yves et al., DNA Topoisomerases and Their Poisoning by Anticancer and Antibacterial Drugs, Chemistry & Biology, May 28, 2010, pp. 421-433, vol. 17, Elsevier Ltd.
Rautio, Jarkko et al., Prodrugs: Design and Clinical Applications, Nature Reviews Drug Discovery, Mar. 2008, pp. 255-270, vol. 7, Nature Publishing Group.
Science (1999), vol. 286, 531-537. *
Staker et al., Structures of Three Classes of Anticancer Agents Bound to the Human Topoisomerase I—DNA Covalent Complex, Journal of Medicinal Chemistry, 2005, pp. 2336-2345, American Chemical Society.
Staker, Bart L. et al., The Mechanism of Topoisomerase I Poisoning by a Campthothecin Analog, Proc. Natl. Acad. Sci, 2002, pp. 15387-15392, vol. 99, No. 24.
Stella, Valentino J., Prodrugs As Therapeutics, Expert Opinion on Therapeutic Patents, 2004, pp. 277-280, vol. 14, No. 3, Taylor & Francis.
Teicher, Beverly A., Next Generation Topoisomerase I Inhibitors: Rationale and Biomarker Strategies, Biochem. Pharmacology, 2008, pp. 1262-1271, vol. 75, Elsevier Inc.
Testa, Bernard, Prodrug Research: Futile or Fertile?, Biochemical Pharmacology, 2004, pp. 2097-2106, vol. 68, Science Direct.
Thomas, Craig J. et al., Camptothecin: Current Perspectives, Bioorganic & Medicinal Chemistry, 2004, pp. 1585-1604, vol. 12, Science Direct.
Wall, Monroe E., et al., Bifunctional Reagents, Cross-linking of Pancreatic Ribonuclease With a Diimido Ester, J. Am. Chem. Soc., 1966, pp. 3888-3890, vol. 88, No. 16.
Wang, Ping, et al. "Synthesis and biological evaluation of the first triple inhibitors of human topoisomerase 1, tyrosyl-DNA phosphodiesterase 1 (Tdp1), and tyrosyl-DNA phosphodiesterase 2 (Tdp2)." Journal of medicinal chemistry 60.8 (2017): 3275-3288.

Also Published As

Publication number Publication date
CA3047992A1 (en) 2018-06-28
EP3558993A4 (en) 2020-05-06
CN110167936A (en) 2019-08-23
JP7182548B2 (en) 2022-12-02
JP2020506886A (en) 2020-03-05
EP3558993A1 (en) 2019-10-30
WO2018118852A1 (en) 2018-06-28
US20200095243A1 (en) 2020-03-26

Similar Documents

Publication Publication Date Title
EP3675839B1 (en) Novel compounds having estrogen receptor alpha degradation activity and uses thereof
KR101586112B1 (en) Carbazole compounds and therapeutic uses of the compounds
CN115335379A (en) Spiro-containing quinazoline compounds
EP4282864A1 (en) Cdk6/dyrk2 dual-target inhibitor, and preparation method therefor and use thereof
WO2007095389A2 (en) Treatment of hyperproliferative diseases with camptothecine n-oxide and analogs
EP3617204A1 (en) Indoleamine 2,3-dioxygenase inhibitor and application
CN113518779B (en) Thieno heterocyclic derivative, preparation method and medical application thereof
US6903133B2 (en) Anticancer compounds
CA3029086C (en) Chiral heterocyclic compound with hedgehog pathway antagonist activity, method and use thereof
JP7245918B2 (en) Sarsasapogenin structure-based derivative, pharmaceutical composition and use thereof
CA3110576A1 (en) Compounds for treating certain leukemias
US10875860B2 (en) Azaindenoisoquinoline compounds and uses thereof
CN107879975A (en) Histon deacetylase (HDAC) inhibitor and its application
EP3284746A1 (en) Preparation and use of kinase inhibitor
KR102606167B1 (en) Fluorine-containing substituted benzothiophene compounds, pharmaceutical compositions and applications thereof
KR102628246B1 (en) Selective A2A receptor antagonist
JP2022554385A (en) WDR5 inhibitors and modulators
US10759795B2 (en) Aza-A-ring indenoisoquinoline topoisomerase I poisons
CN104341407A (en) Quinazoline compounds, preparation method and applications thereof
US20230312562A1 (en) Tetrahydro-3h-pyrazolo quinolone and tetrahydro-3h-pyrrolo[3,2-f]quinoline -containing compounds and uses thereof
US20190218226A1 (en) Prodrugs of anticancer agents indotecan and indimitecan
CN117384162A (en) Selective HER2 inhibitors
CN116669741A (en) Polycyclic kinase inhibitors
CN117658983A (en) Selective PARP1 inhibitors
CN116848117A (en) Fused ring compounds as Wee-1 inhibitors

Legal Events

Date Code Title Description
FEPP Fee payment procedure

Free format text: ENTITY STATUS SET TO UNDISCOUNTED (ORIGINAL EVENT CODE: BIG.); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

AS Assignment

Owner name: PURDUE RESEARCH FOUNDATION, INDIANA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:CUSHMAN, MARK S.;WANG, PING;ELSAYED, MOHAMED S.A.;SIGNING DATES FROM 20171207 TO 20171212;REEL/FRAME:050343/0116

Owner name: PURDUE RESEARCH FOUNDATION, INDIANA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:CUSHMAN, MARK S;WANG, PING;ELSAYED, MOHAMED S.A.;SIGNING DATES FROM 20171207 TO 20171212;REEL/FRAME:050343/0072

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NOTICE OF ALLOWANCE MAILED -- APPLICATION RECEIVED IN OFFICE OF PUBLICATIONS

AS Assignment

Owner name: THE UNITED STATES OF AMERICA, AS REPRESENTED BY THE SECRETARY, DEPARTMENT OF HEALTH AND HUMAN SERVICES, MARYLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:POMMIER, YVES G.;REEL/FRAME:053899/0883

Effective date: 20200928

STPP Information on status: patent application and granting procedure in general

Free format text: PUBLICATIONS -- ISSUE FEE PAYMENT VERIFIED

STCF Information on status: patent grant

Free format text: PATENTED CASE

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 4TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1551); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 4